1
|
Chiu CH, Lu CW, Lin TY, Chen PF. Abdominal compartment syndrome during intraoperative hyperthermic intraperitoneal chemotherapy following debulking surgery for colorectal cancer: A case report. Int J Surg Case Rep 2024; 123:110192. [PMID: 39241478 PMCID: PMC11405813 DOI: 10.1016/j.ijscr.2024.110192] [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: 06/24/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/09/2024] Open
Abstract
INTRODUCTION Colorectal cancer leads to peritoneal metastasis in 8-15 % of cases and necessitates treatments, such as hyperthermic intraperitoneal chemotherapy (HIPEC). However, HIPEC may result in perioperative complications, some often overlooked, such as abdominal compartment syndrome. CASE PRESENTATION A 52-year-old female with colorectal cancer and peritoneal metastasis underwent debulking surgery followed by HIPEC. During HIPEC, a sudden increase in airway pressure and severe hypotension were noted. Pneumothorax with abdominal compartment syndrome (ACS) was suspected and HIPEC was terminated. Despite intravenous fluids and vasopressors, she experienced circulatory and respiratory collapse. Laparotomy sutures were promptly removed, which effectively alleviated the intra-abdominal hypertension and immediately restored the vital signs. An inadequately repaired diaphragm defect was identified and repaired. A chest tube was inserted for pleural effusion. DISCUSSION ACS is characterized by an increase in abdominal cavity pressure above 20 mmHg, leading to end-organ damage. It can mimic physiological effects of HIPEC and result in adverse outcomes. Early detection of ACS is essential, especially when complicated by pneumothorax from diaphragmatic tumor dissection. The closed technique for HIPEC, while efficient, can increase the risk of ACS and requires careful management. CONCLUSIONS This case underscores the complexity of HIPEC and the importance of promptly identifying and managing ACS during the procedure. Monitoring intra-abdominal pressure during HIPEC is essential. Thoroughly check for iatrogenic injuries, including the diaphragm, is crucial before starting before HIPEC.
Collapse
Affiliation(s)
- Chen-Hsi Chiu
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Cheng-Wei Lu
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan 320, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan 320, Taiwan
| | - Pei-Fu Chen
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan 320, Taiwan.
| |
Collapse
|
2
|
Pisuchpen N, Lennartz S, Parakh A, Kongboonvijit S, Srinivas Rao S, Pierce TT, Anderson MA, Hahn PF, Mercaldo ND, Kambadakone A. Material density dual-energy CT images: value added in early diagnosis of peritoneal carcinomatosis : Original research. Abdom Radiol (NY) 2024; 49:3496-3506. [PMID: 38916617 DOI: 10.1007/s00261-024-04451-0] [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: 04/11/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
OBJECTIVE To assess the value of material density (MD) images generated from a rapid kilovoltage-switching dual-energy CT (rsDECT) in early detection of peritoneal carcinomatosis (PC). MATERIALS AND METHODS Thirty patients (60 ± 13 years; 24 women) with PC detected on multiple abdominal DECT scans were included. Four separate DECTs with varying findings of PC from each patient were used for qualitative/quantitative analysis, resulting in a total of 120 DECT scans (n = 30 × 4). Three radiologists independently reviewed DECT images (65 keV alone and 65 keV + MD) for diagnosis of PC (diagnostic confidence, lesion conspicuity, sharpness/delineation and image quality) using a 5-point Likert scale. Quantitative estimation of contrast-to-noise ratio (CNR) was done. Wilcoxon signed-rank test and Odds ratio calculation were used to compare between the two protocols. Inter-observer agreement was evaluated using Kappa coefficient analysis. P values < 0.05 were considered statistically significant. RESULTS 65 keV + MD images showed a slightly higher sensitivity (89%[95%CI:84,92]) for PC detection compared with 65 keV images alone without statistical significance (84%[95%CI:78,88], p = 0.11) with the experienced reader showing significant improvement (98%[95%CI:93,100] vs. 90%[95%CI:83,94], p = 0.02). On a per-patient basis, use of MD images allowed earlier diagnosis for PC in an additional 13-23% of patients. On sub-group analysis, earlier diagnosis of PC was particularly beneficial in patients with BMI ≤ 29.9 kg/m2. 65 keV + MD images showed higher diagnostic confidence, lesion conspicuity, and lesion sharpness for the experienced reader (p < 0.001). CNR was higher in MD images (1.7 ± 0.5) than 65 keV images (0.1 ± 0.02, p < 0.001). All readers showed moderate interobserver agreement for determining PC by both protocols (κ = 0.58 and κ = 0.47). CONCLUSION MD images allow earlier and improved detection of PC with the degree of benefit varying based on reader experience and patient body habitus.
Collapse
Affiliation(s)
- Nisanard Pisuchpen
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
- Department of Radiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Simon Lennartz
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University Cologne, 50937, Cologne, Germany
| | - Anushri Parakh
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Sasiprang Kongboonvijit
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
- Department of Radiology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Shravya Srinivas Rao
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Theodore T Pierce
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Mark A Anderson
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Peter F Hahn
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Nathaniel D Mercaldo
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Avinash Kambadakone
- Abdominal Radiology Division, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, White 270, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Mc Larney BE, Sonay AY, Apfelbaum E, Mostafa N, Monette S, Goerzen D, Aguirre N, Exner RM, Habjan C, Isaac E, Phung NB, Skubal M, Kim M, Ogirala A, Veach D, Heller DA, Grimm J. A pan-cancer dye for solid-tumour screening, resection and wound monitoring via short-wave and near-infrared fluorescence imaging. Nat Biomed Eng 2024; 8:1092-1108. [PMID: 39251765 DOI: 10.1038/s41551-024-01248-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/21/2024] [Indexed: 09/11/2024]
Abstract
The efficacy of fluorescence-guided surgery in facilitating the real-time delineation of tumours depends on the optical contrast of tumour tissue over healthy tissue. Here we show that CJ215-a commercially available, renally cleared carbocyanine dye sensitive to apoptosis, and with an absorption and emission spectra suitable for near-infrared fluorescence imaging (wavelengths of 650-900 nm) and shortwave infrared (SWIR) fluorescence imaging (900-1,700 nm)-can facilitate fluorescence-guided tumour screening, tumour resection and the assessment of wound healing. In tumour models of either murine or human-derived breast, prostate and colon cancers and of fibrosarcoma, and in a model of intraperitoneal carcinomatosis, imaging of CJ215 with ambient light allowed for the delineation of nearly all tumours within 24 h after intravenous injection of the dye, which was minimally taken up by healthy organs. At later timepoints, CJ215 provided tumour-to-muscle contrast ratios up to 100 and tumour-to-liver contrast ratios up to 18. SWIR fluorescence imaging with the dye also allowed for quantifiable non-contact wound monitoring through commercial bandages. CJ215 may be compatible with existing and emerging clinical solutions.
Collapse
Affiliation(s)
| | - Ali Yasin Sonay
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elana Apfelbaum
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA
| | - Nermin Mostafa
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sébastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, NY, USA
| | - Dana Goerzen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Nicole Aguirre
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rüdiger M Exner
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christine Habjan
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA
| | - Elizabeth Isaac
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA
| | - Ngan Bao Phung
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA
| | - Magdalena Skubal
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mijin Kim
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Anuja Ogirala
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Darren Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical Center, New York, NY, USA
| | - Daniel A Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jan Grimm
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical Center, New York, NY, USA.
- Molecular Imaging Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Bohosova J, Ashraf NS, Slaby O, Calin GA. Non-Coding RNAs in Peritoneal Carcinomatosis: From Bench to Bedside. Cancers (Basel) 2024; 16:2961. [PMID: 39272819 PMCID: PMC11394633 DOI: 10.3390/cancers16172961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Peritoneal carcinomatosis represents an advanced stage of tumors within the peritoneal cavity. Once considered an incurable terminal cancer metastasis, contemporary medicine is on the hunt for certain potentially curative options alongside the present day's palliative disease management. However, for most patients, peritoneal carcinomatosis continues to pose a fatal late-stage prognosis with a grim future outlook. Over the past two decades, non-coding RNAs have garnered significant attention due to their undeniable significance in regulating cellular processes across all levels. Disruption of the intricate regulation led by non-coding RNAs has been demonstrated to have a substantial impact on various human diseases, particularly in cancer, including solid tumors originating from the organs of the peritoneal cavity. This review aims to offer a comprehensive overview of the current state of knowledge in the under-researched field of peritoneal carcinomatosis, focusing specifically on the role of non-coding RNAs in the development of this condition and delineating potential avenues for future research.
Collapse
Affiliation(s)
- Julia Bohosova
- Translational Molecular Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Nida Sarosh Ashraf
- Translational Molecular Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Islamabad 45550, Pakistan
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - George A Calin
- Translational Molecular Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The RNA Interference and Non-Coding RNA Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Khalafi S, Botero Fonnegra C, Reyes A, Hui VW. Developments in the Use of Indocyanine Green (ICG) Fluorescence in Colorectal Surgery. J Clin Med 2024; 13:4003. [PMID: 39064041 PMCID: PMC11276973 DOI: 10.3390/jcm13144003] [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: 05/20/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Indocyanine Green (ICG) has significantly advanced minimally invasive surgery. It is widely recognized for its ability to visualize blood vessel patency in real-time across various surgical specialties. While its primary use in colorectal surgery is to evaluate anastomoses for leaks, numerous other applications have been documented in the literature. In this review, we aim to explore both established and emerging applications of ICG fluorescence in colorectal surgery, with the goal of improving patient outcomes. This includes preoperative tumor marking and the detection of metastatic disease. Some applications, such as lymphatic mapping, require further research to determine their impact on clinical practices. Conversely, others, like the intraoperative localizations of ureters, necessitate additional procedures and are not yet widely accepted by the surgical community. However, the development of alternative compounds could offer better solutions. Future research should focus on areas like quantitative ICG and protocol standardization in prospective multicenter studies.
Collapse
|
6
|
Min SH, Lee J, Yoo M, Hwang D, Lee E, Kang SH, Lee K, Park YS, Ahn SH, Suh YS, Park DJ, Kim HH. Efficacy of Hyperthermic Pressurized Intraperitoneal Aerosol Chemotherapy in an In Vitro Model Using a Human Gastric Cancer AGS Cell Line and an Abdominal Cavity Model. J Gastric Cancer 2024; 24:246-256. [PMID: 38960884 PMCID: PMC11224724 DOI: 10.5230/jgc.2024.24.e24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE Peritoneal carcinomatosis (PC) presents a major challenge in the treatment of late-stage, solid tumors, with traditional therapies limited by poor drug penetration. We evaluated a novel hyperthermic pressurized intraperitoneal aerosol chemotherapy (HPIPAC) system using a human abdominal cavity model for its efficacy against AGS gastric cancer cells. MATERIALS AND METHODS A model simulating the human abdominal cavity and AGS gastric cancer cell line cultured dishes were used to assess the efficacy of the HPIPAC system. Cell viability was measured to evaluate the impact of HPIPAC under 6 different conditions: heat alone, PIPAC with paclitaxel (PTX), PTX alone, normal saline (NS) alone, heat with NS, and HPIPAC with PTX. RESULTS Results showed a significant reduction in cell viability with HPIPAC combined with PTX, indicating enhanced cytotoxic effects. Immediately after treatment, the average cell viability was 66.6%, which decreased to 49.2% after 48 hours and to a further 19.6% after 120 hours of incubation, demonstrating the sustained efficacy of the treatment. In contrast, control groups exhibited a recovery in cell viability; heat alone showed cell viability increasing from 90.8% to 94.4%, PIPAC with PTX from 82.7% to 89.7%, PTX only from 73.3% to 74.8%, NS only from 90.9% to 98.3%, and heat with NS from 74.4% to 84.7%. CONCLUSIONS The HPIPAC system with PTX exhibits a promising approach in the treatment of PC in gastric cancer, significantly reducing cell viability. Despite certain limitations, this study highlights the system's potential to enhance treatment outcomes. Future efforts should focus on refining HPIPAC and validating its effectiveness in clinical settings.
Collapse
Affiliation(s)
- Sa-Hong Min
- Department of Gastrointestinal Surgery, Asan Medical Center, Seoul, Korea
| | - Jieun Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Mira Yoo
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Duyeong Hwang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Eunju Lee
- Department of Surgery, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Korea
| | - So Hyun Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kanghaeng Lee
- Department of Surgery, St. Vincent's Hospital, Suwon, Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
7
|
Taqi K, Lee J, Hurton S, Stockley C, Mack L, Rivard J, Temple W, Bouchard-Fortier A. Long-Term Outcomes following Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Peritoneal Carcinomatosis of Colorectal Origin. Curr Oncol 2024; 31:3657-3668. [PMID: 39057141 PMCID: PMC11275434 DOI: 10.3390/curroncol31070269] [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: 05/22/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is a major treatment of colorectal peritoneal carcinomatosis (CPC). The aim was to determine the disease-free survival (DFS) and overall survival (OS) of patients undergoing CRS-HIPEC for CPC and factors associated with long-term survival (LTS). METHODS consecutive CPC patients who underwent CRS-HIPEC at a HIPEC center between 2007 and 2021 were included. Actual survival was calculated, and Cox proportional hazards models were used to identify factors associated with OS, DFS and LTS. RESULTS there were 125 patients with CPC who underwent primary CRS-HIPEC, with mean age of 54.5 years. Median follow-up was 31 months. Average intraoperative PCI was 11, and complete cytoreduction (CC-0) was achieved in 96.8%. Median OS was 41.6 months (6-196). The 2-year and 5-year OS were 68% and 24.8%, respectively, and the 2-year DFS was 28.8%. Factors associated with worse OS included pre-HIPEC systemic therapy, synchronous extraperitoneal metastasis, and PCI ≥ 20 (p < 0.05). Progression prior to CRS-HIPEC was associated with worse DFS (p < 0.05). Lower PCI, fewer complications, lower recurrence and longer DFS were associated with LTS (p < 0.05). CONCLUSION CRS and HIPEC improve OS in CPC patients but they have high disease recurrence. Outcomes depend on preoperative therapy response, extraperitoneal metastasis, and peritoneal disease burden.
Collapse
Affiliation(s)
- Kadhim Taqi
- Division of Surgical Oncology, Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jay Lee
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Scott Hurton
- Division of Surgery, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Cecily Stockley
- Division of Surgical Oncology, Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Lloyd Mack
- Division of Surgical Oncology, Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Justin Rivard
- Division of Surgery, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Walley Temple
- Division of Surgical Oncology, Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Antoine Bouchard-Fortier
- Division of Surgical Oncology, Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
8
|
Wei GX, Zhou YW, Li ZP, Qiu M. Application of artificial intelligence in the diagnosis, treatment, and recurrence prediction of peritoneal carcinomatosis. Heliyon 2024; 10:e29249. [PMID: 38601686 PMCID: PMC11004411 DOI: 10.1016/j.heliyon.2024.e29249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Peritoneal carcinomatosis (PC) is a type of secondary cancer which is not sensitive to conventional intravenous chemotherapy. Treatment strategies for PC are usually palliative rather than curative. Recently, artificial intelligence (AI) has been widely used in the medical field, making the early diagnosis, individualized treatment, and accurate prognostic evaluation of various cancers, including mediastinal malignancies, colorectal cancer, lung cancer more feasible. As a branch of computer science, AI specializes in image recognition, speech recognition, automatic large-scale data extraction and output. AI technologies have also made breakthrough progress in the field of peritoneal carcinomatosis (PC) based on its powerful learning capacity and efficient computational power. AI has been successfully applied in various approaches in PC diagnosis, including imaging, blood tests, proteomics, and pathological diagnosis. Due to the automatic extraction function of the convolutional neural network and the learning model based on machine learning algorithms, AI-assisted diagnosis types are associated with a higher accuracy rate compared to conventional diagnosis methods. In addition, AI is also used in the treatment of peritoneal cancer, including surgical resection, intraperitoneal chemotherapy, systemic chemotherapy, which significantly improves the survival of patients with PC. In particular, the recurrence prediction and emotion evaluation of PC patients are also combined with AI technology, further improving the quality of life of patients. Here we have comprehensively reviewed and summarized the latest developments in the application of AI in PC, helping oncologists to comprehensively diagnose PC and provide more precise treatment strategies for patients with PC.
Collapse
Affiliation(s)
- Gui-Xia Wei
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Zhi-Ping Li
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Arrizabalaga L, Di Trani CA, Risson A, Belsúe V, Gomar C, Ardaiz N, Berrondo P, Aranda F, Bella Á. Peritoneal carcinomatosis in mouse models. Methods Cell Biol 2024; 185:67-78. [PMID: 38556452 DOI: 10.1016/bs.mcb.2024.02.005] [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] [Indexed: 04/02/2024]
Abstract
Peritoneal carcinomatosis (PCa) represents a metastatic stage of a disease with unmet therapeutic options. Malignant cells from primary tumors (gastrointestinal or gynecologic malignancies) invade the peritoneal cavity and eventually seed onto peritoneal surfaces, with the omentum being the most common nest area. With a median survival of less than 6 months, PCa has a dismal prognosis that can be improved with treatments only available to a select few individuals with low tumor burden. Thus, the discovery of novel and effective therapies for this disease depends on reliable animal models. Here, we describe a method to generate syngeneic PCa mouse models based on intraperitoneal (i.p.) administration of tumor cells. This model allows to follow-up cancer progression in PCa models from ovarian and colorectal origins monitoring mice bodyweight changes, ascites development and overall survival. Moreover, luciferase-expressing tumor cells can also be used to assess tumor growth after i.p. injection through in vivo bioluminescence quantification. The establishment of reliable, easy-to-monitor and reproducible intraperitoneal syngeneic tumors models, as described here, is the first step to develop cutting-edge therapies against PCa.
Collapse
Affiliation(s)
- Leire Arrizabalaga
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
| | - Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Aline Risson
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Virginia Belsúe
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Celia Gomar
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Nuria Ardaiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Pedro Berrondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
| | - Ángela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
| |
Collapse
|
10
|
Ilyas MIM. Epidemiology of Stage IV Colorectal Cancer: Trends in the Incidence, Prevalence, Age Distribution, and Impact on Life Span. Clin Colon Rectal Surg 2024; 37:57-61. [PMID: 38322602 PMCID: PMC10843881 DOI: 10.1055/s-0043-1761447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Colorectal cancer is a common malignancy in men and women. Historically, stage IV colorectal cancer has 10 to 15% five-year survival. Developments in the management of colorectal metastatic disease have helped improve the overall survival of stage IV colorectal cancers from 12 to 30 months with some patients achieving disease-free survival.
Collapse
|
11
|
Mc Larney B, Sonay A, Apfelbaum E, Mostafa N, Monette S, Goerzen D, Aguirre N, Isaac E, Phung N, Skubal M, Kim M, Ogirala A, Veach D, Heller D, Grimm J. A pan-cancer agent for screening, resection and wound monitoring via NIR and SWIR imaging. RESEARCH SQUARE 2024:rs.3.rs-3879635. [PMID: 38343820 PMCID: PMC10854300 DOI: 10.21203/rs.3.rs-3879635/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Fluorescence guided surgery (FGS) facilitates real time tumor delineation and is being rapidly established clinically. FGS efficacy is tied to the utilized dye and provided tumor contrast over healthy tissue. Apoptosis, a cancer hallmark, is a desirable target for tumor delineation. Here, we preclinically in vitro and in vivo, validate an apoptosis sensitive commercial carbocyanine dye (CJ215), with absorption and emission spectra suitable for near infrared (NIR, 650-900nm) and shortwave infrared (SWIR, 900-1700nm) fluorescence imaging (NIRFI, SWIRFI). High contrast SWIRFI for solid tumor delineation is demonstrated in multiple murine and human models including breast, prostate, colon, fibrosarcoma and intraperitoneal colorectal metastasis. Organ necropsy and imaging highlighted renal clearance of CJ215. SWIRFI and CJ215 delineated all tumors under ambient lighting with a tumor-to-muscle ratio up to 100 and tumor-to-liver ratio up to 18, from 24 to 168 h post intravenous injection with minimal uptake in healthy organs. Additionally, SWIRFI and CJ215 achieved non-contact quantifiable wound monitoring through commercial bandages. CJ215 provides tumor screening, guided resection, and wound healing assessment compatible with existing and emerging clinical solutions.
Collapse
Affiliation(s)
| | - Ali Sonay
- Memorial Sloan Kettering Cancer Center
| | | | | | | | | | | | | | | | | | - Mijin Kim
- Memorial Sloan Kettering Cancer Center
| | | | | | | | - Jan Grimm
- Memorial Sloan Kettering Cancer Center
| |
Collapse
|
12
|
Berlin C, Mauerer B, Cauchy P, Luenstedt J, Sankowski R, Marx L, Feuerstein R, Schaefer L, Greten FR, Pesic M, Groß O, Prinz M, Ruehl N, Miketiuk L, Jauch D, Laessle C, Jud A, Biesel EA, Neeff H, Fichtner-Feigl S, Holzner PA, Kesselring R. Single-cell deconvolution reveals high lineage- and location-dependent heterogeneity in mesenchymal multivisceral stage 4 colorectal cancer. J Clin Invest 2023; 134:e169576. [PMID: 38153787 PMCID: PMC10904044 DOI: 10.1172/jci169576] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023] Open
Abstract
Metastasized colorectal cancer (CRC) is associated with a poor prognosis and rapid disease progression. Besides hepatic metastasis, peritoneal carcinomatosis is the major cause of death in Union for International Cancer Control (UICC) stage IV CRC patients. Insights into differential site-specific reconstitution of tumor cells and the corresponding tumor microenvironment are still missing. Here, we analyzed the transcriptome of single cells derived from murine multivisceral CRC and delineated the intermetastatic cellular heterogeneity regarding tumor epithelium, stroma, and immune cells. Interestingly, we found an intercellular site-specific network of cancer-associated fibroblasts and tumor epithelium during peritoneal metastasis as well as an autologous feed-forward loop in cancer stem cells. We furthermore deciphered a metastatic dysfunctional adaptive immunity by a loss of B cell-dependent antigen presentation and consecutive effector T cell exhaustion. Furthermore, we demonstrated major similarities of this murine metastatic CRC model with human disease and - based on the results of our analysis - provided an auspicious site-specific immunomodulatory treatment approach for stage IV CRC by intraperitoneal checkpoint inhibition.
Collapse
Affiliation(s)
- Christopher Berlin
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- IMM-PACT Clinician Scientist Program
| | - Bernhard Mauerer
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pierre Cauchy
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jost Luenstedt
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- IMM-PACT Clinician Scientist Program
| | - Roman Sankowski
- Institute of Neuropathology
- Single-Cell Omics Platform Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lisa Marx
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Reinhild Feuerstein
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Luisa Schaefer
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian R. Greten
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Marina Pesic
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Olaf Groß
- Institute of Neuropathology
- Signalling Research Centres BIOSS and CIBSS
| | - Marco Prinz
- Institute of Neuropathology
- Signalling Research Centres BIOSS and CIBSS
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, and
| | - Naomi Ruehl
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Miketiuk
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dominik Jauch
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Laessle
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- EXCEL Excellent Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Jud
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Esther A. Biesel
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hannes Neeff
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp A. Holzner
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rebecca Kesselring
- Department of General and Visceral Surgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
13
|
Wang M, Hu D, Yang Y, Shi K, Li J, Liu Q, Li Y, Li R, Pan M, Mo D, Chen W, Li X, Qian Z. Enhanced Chemo-Immunotherapy Strategy Utilizing Injectable Thermosensitive Hydrogel for The Treatment of Diffuse Peritoneal Metastasis in Advanced Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303819. [PMID: 37875399 PMCID: PMC10724414 DOI: 10.1002/advs.202303819] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/17/2023] [Indexed: 10/26/2023]
Abstract
Patients with colorectal cancer (CRC) and diffuse peritoneal metastasis (PM) are not eligible for surgical intervention. Thus, palliative treatment remains the standard of care in clinical practice. Systemic chemotherapy fails to cause drug accumulation at the lesion sites, while intraperitoneal chemotherapy (IPC) is limited by high clearance rates and associated complications. Given the poor prognosis, a customized OxP/R848@PLEL hydrogel delivery system has been devised to improve the clinical benefit of advanced CRC with diffuse PM. This system is distinguished by its simplicity, security, and efficiency. Specifically, the PLEL hydrogel exhibits excellent injectability and thermosensitivity, enabling the formation of drug depots within the abdominal cavity, rendering it an optimal carrier for IPC. Oxaliplatin (OxP), a first-line drug for advanced CRC, is cytotoxic and enhances the immunogenicity of tumors by inducing immunogenic cell death. Furthermore, OxP and resiquimod (R848) synergistically enhance the maturation of dendritic cells, promote the expansion of cytotoxic T lymphocytes, and induce the formation of central memory T cells. Moreover, R848 domesticates macrophages to an anti-tumor phenotype. OxP/R848@PLEL effectively eradicates peritoneal metastases, completely inhibits ascites production, and significantly prolongs mice lifespan. As such, it provides a promising approach to managing diffuse PM in patients with CRC without surgical indications.
Collapse
Affiliation(s)
- Meng Wang
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - DanRong Hu
- Rehabilitation Medicine Center and Institute of Rehabilitation MedicineKey Laboratory of Rehabilitation Medicine in Sichuan ProvinceWest China HospitalSichuan UniversityChengdu610041China
| | - Yun Yang
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Kun Shi
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - JiaNan Li
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - QingYa Liu
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - YiCong Li
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Ran Li
- Rehabilitation Medicine Center and Institute of Rehabilitation MedicineKey Laboratory of Rehabilitation Medicine in Sichuan ProvinceWest China HospitalSichuan UniversityChengdu610041China
| | - Meng Pan
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Dong Mo
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Wen Chen
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - XiCheng Li
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - ZhiYong Qian
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
14
|
Bitsianis S, Mantzoros I, Anestiadou E, Christidis P, Chatzakis C, Zapsalis K, Symeonidis S, Ntampakis G, Domvri K, Tsakona A, Bekiari C, Ioannidis O, Aggelopoulos S. Effect of Intraperitoneal Chemotherapy with Regorafenib on IL-6 and TNF-α Levels and Peritoneal Cytology: Experimental Study in Rats with Colorectal Peritoneal Carcinomatosis. J Clin Med 2023; 12:7267. [PMID: 38068319 PMCID: PMC10706907 DOI: 10.3390/jcm12237267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 08/11/2024] Open
Abstract
Cytoreductive surgery (CRS), combined with hyperthermic intraperitoneal chemotherapy, has significantly improved survival outcomes in patients with peritoneal carcinomatosis from colorectal cancer (CRC). Regorafenib is an oral agent administered in patients with refractory metastatic CRC. Our aim was to investigate the outcomes of intraperitoneal administration of regorafenib for intraperitoneal chemotherapy (IPEC) or/and CRS in a rat model of colorectal peritoneal metastases regarding immunology and peritoneal cytology. A total of 24 rats were included. Twenty-eight days after carcinogenesis induction, rats were randomized into following groups: group A: control group; group B: CRS only; group C: IPEC only; and group D: CRS + IPEC. On day 56 after carcinogenesis, euthanasia and laparotomy were performed. Serum levels of interleukin-6 (IL-6) and tumor necrosis factor α (TNF-α) as well as peritoneal cytology were investigated. Groups B and D had statistically significant lower mean levels of IL-6 and TNF-α compared to groups A and C, but there was no significant difference between them. Both B and D groups presented a statistically significant difference regarding the rate of negative peritoneal cytology, when compared to the control group, but not to group C. In conclusion, regorafenib-based IPEC, combined with CRS, may constitute a promising tool against peritoneal carcinomatosis by altering the tumor microenvironment.
Collapse
Affiliation(s)
- Stefanos Bitsianis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Ioannis Mantzoros
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Elissavet Anestiadou
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Panagiotis Christidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Christos Chatzakis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Konstantinos Zapsalis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Savvas Symeonidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Georgios Ntampakis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Kalliopi Domvri
- Laboratory of Histology-Embryology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Pathology, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece
| | - Anastasia Tsakona
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Chryssa Bekiari
- Experimental and Research Center, Papageorgiou General Hospital of Thessaloniki, 56403 Thessaloniki, Greece;
- Laboratory of Anatomy and Histology, Veterinary School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Orestis Ioannidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| | - Stamatios Aggelopoulos
- 4th Department of General Surgery, Aristotle University of Thessaloniki, General Hospital of Thessaloniki “G. Papanikolaou”, 54124 Thessaloniki, Greece; (S.B.); (I.M.); (E.A.); (P.C.); (C.C.); (K.Z.); (S.S.); (G.N.); (S.A.)
| |
Collapse
|
15
|
Heuvelings DJI, Wintjens AGWE, Moonen L, Engelen SME, de Hingh IHJT, Valkenburg-van Iersel LB, den Dulk M, Beckervordersandforth J, Thijssen SGM, Leunissen DJG, Stassen LPS, Keszthelyi D, Mujagic Z, Speel EJM, Bouvy ND. Predictive Genetic Biomarkers for the Development of Peritoneal Metastases in Colorectal Cancer. Int J Mol Sci 2023; 24:12830. [PMID: 37629011 PMCID: PMC10454220 DOI: 10.3390/ijms241612830] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Metastatic colorectal cancer (CRC) is a common cause of cancer-related mortality, of which peritoneal metastases (PMs) have the worse outcome. Metastasis-specific markers may help predict the spread of tumor cells and select patients for preventive strategies. This exploratory pilot study aimed to gain more insight into genetic alterations in primary CRC tumors, which might be a predictive factor for the development of PM. Forty patients with T3 stage CRC were retrospectively divided in three groups: without metachronous metastases during 5-year follow-up (M0, n = 20), with metachronous liver metastases (LM, n = 10) and with metachronous PM (PM, n = 10). Patients with synchronous metastases were excluded. Primary formalin-fixed paraffin-embedded tumor samples were analyzed via comprehensive genome sequencing (TSO500 analysis) to identify DNA alterations and RNA fusion transcripts in 523 genes and 55 genes, respectively. Thirty-eight samples were included for final analysis. Four M0 tumors and one PM tumor were microsatellite instable. BRAF mutations were uniquely identified in three microsatellite-stable (MSS) PM tumors (37.5%, p = 0.010). RNA analysis showed an additional FAM198A-RAF1 fusion in one PM sample. BRAF p.V600E mutations were only present in PM patients with MSS tumors. Greater attention should be paid to BRAF-mutated tumors in relation to the development of metachronous PM.
Collapse
Affiliation(s)
- Danique J. I. Heuvelings
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Anne G. W. E. Wintjens
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Laura Moonen
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Sanne M. E. Engelen
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Ignace H. J. T. de Hingh
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Catharina Ziekenhuis, 5623 EJ Eindhoven, The Netherlands
| | - Liselot B. Valkenburg-van Iersel
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Internal Medicine, Division of Medical Oncology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Marcel den Dulk
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Jan Beckervordersandforth
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Sharon G. M. Thijssen
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Daphne J. G. Leunissen
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Laurents P. S. Stassen
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Daniel Keszthelyi
- Division of Gastroenterology and Hepatology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Zlatan Mujagic
- Division of Gastroenterology and Hepatology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Ernst-Jan M. Speel
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Nicole D. Bouvy
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
16
|
Shin W, Yun J, Han K, Park DG. Comparison of genetic variation between primary colorectal cancer and metastatic peritoneal cancer. Genes Genomics 2023; 45:989-1001. [PMID: 37277571 DOI: 10.1007/s13258-023-01408-3] [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: 05/04/2023] [Accepted: 05/21/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND Among cancer metastases by primary colorectal cancer (CRC), peritoneal metastasis is the second most common metastatic lesion after liver metastasis. In treating metastatic CRC, it is very important to differentiate targeted-therapy and chemotherapy according to the characteristics of each lesion because the genetic variation of the primary and metastatic lesions are different. However, there are few studies of genetic characteristics on peritoneal metastasis caused by primary CRC, so molecular-level studies are continuously required. OBJECTIVE We propose an appropriate peritoneal metastasis treatment policy by identifying the genetic characteristics between primary CRC and synchronous peritoneal metastatic lesions. METHODS Primary CRC and synchronous peritoneal metastasis samples were analyzed in pairs from six patients using Comprehensive Cancer Panel (409 cancer-related genes, Thermo Fisher Scientific, USA) and next-generation sequencing (NGS). RESULTS The mutations were commonly found on the KMT2C and THBS1 genes in both primary CRC and peritoneal metastasis. The PDE4DIP gene was mutated in all cases except for on a sample of peritoneal metastasis. As a result of analysis using the mutation database, we confirmed that the gene mutations of primary CRC and the peritoneal metastasis derived from it showed the same tendency, although we did not accompany the gene expression level or epigenetic study. CONCLUSION It is thought that the treatment policy through molecular genetic testing of primary CRC can also be applied to peritoneal metastasis treatment. Our study is expected to be the basis for further peritoneal metastasis research.
Collapse
Affiliation(s)
- Wonseok Shin
- NGS Clinical Laboratory, Dankook University Hospital, Cheonan, Republic of Korea
| | - Jeongseok Yun
- Department of Surgery, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Kyudong Han
- NGS Clinical Laboratory, Dankook University Hospital, Cheonan, Republic of Korea.
- Department of Microbiology, Dankook University, Cheonan, Republic of Korea.
- Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan, Republic of Korea.
- R&D Center, HuNbiome Co., Ltd, Seoul, Republic of Korea.
| | - Dong-Guk Park
- NGS Clinical Laboratory, Dankook University Hospital, Cheonan, Republic of Korea.
- Department of Surgery, Dankook University College of Medicine, Cheonan, Republic of Korea.
| |
Collapse
|
17
|
Heuvelings DJI, Wintjens AGWE, Luyten J, Wilmink GEWA, Moonen L, Speel EJM, de Hingh IHJT, Bouvy ND, Peeters A. DNA and RNA Alterations Associated with Colorectal Peritoneal Metastases: A Systematic Review. Cancers (Basel) 2023; 15:cancers15020549. [PMID: 36672497 PMCID: PMC9856984 DOI: 10.3390/cancers15020549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND As colorectal cancer (CRC) patients with peritoneal metastases (PM) have a poor prognosis, new treatment options are currently being investigated for CRC patients. Specific biomarkers in the primary tumor could serve as a prediction tool to estimate the risk of distant metastatic spread. This would help identify patients eligible for early treatment. AIM To give an overview of previously studied DNA and RNA alterations in the primary tumor correlated to colorectal PM and investigate which gene mutations should be further studied. METHODS A systematic review of all published studies reporting genomic analyses on the primary tissue of CRC tumors in relation to PM was undertaken according to PRISMA guidelines. RESULTS Overall, 32 studies with 18,906 patients were included. BRAF mutations were analyzed in 17 articles, of which 10 found a significant association with PM. For all other reported genes, no association with PM was found. Two analyses with broader cancer panels did not reveal any new biomarkers. CONCLUSION An association of specific biomarkers in the primary tumors of CRC patients with metastatic spread into peritoneum could not be proven. The role of BRAF mutations should be further investigated. In addition, studies searching for potential novel biomarkers are still required.
Collapse
Affiliation(s)
- Danique J. I. Heuvelings
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence:
| | - Anne G. W. E. Wintjens
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Julien Luyten
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
| | - Guus E. W. A. Wilmink
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Laura Moonen
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ernst-Jan M. Speel
- Department of Pathology, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ignace H. J. T. de Hingh
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General Surgery, Catharina Ziekenhuis, 5623 EJ Eindhoven, The Netherlands
| | - Nicole D. Bouvy
- Department of General Surgery, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands
- GROW–School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Andrea Peeters
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre (MUMC+), 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
18
|
Alhumaidan SS, Alharbi AM, Syeda AF, Alghaidani FA, Almutairi MM, Alharbi NA, Alenezi RK. Hyperthermic Intraperitoneal Chemotherapy Following Cytoreductive Surgery for Colorectal Peritoneal Carcinomatosis Patients: A Review. Cureus 2022; 14:e32440. [PMID: 36523856 PMCID: PMC9744358 DOI: 10.7759/cureus.32440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2022] [Indexed: 12/14/2022] Open
Abstract
Colorectal peritoneal carcinomatosis (CPC) is an advanced malignancy and is typically associated with a poor prognosis. Hyperthermic intraperitoneal chemotherapy (HIPEC) following complete cytoreductive surgery (CRS) is a novel, advanced loco-regional treatment for colorectal cancer that is currently being used to treat peritoneal carcinomatosis (PC). The present review aims to describe the evidence-based literature on the efficacy and safety of this treatment approach in patients with PC originating from colorectal cancer and to summarize its complications. All published literature regarding the efficacy of HIPEC for the treatment of CPC was reviewed; 16 studies were included in this paper. The overall survival rate for the HIPEC group ranged from 63% to 93%. The overall median survival for the HIPEC and non-HIPEC groups ranged from 13 to 60.1 months and 12.6 to 41.2 months, respectively. The overall median survival of patients in the HIPEC group was comparatively better than those in the non-HIPEC group. There was insufficient evidence to suggest whether this treatment regimen was associated with a high or low morbidity rate in comparison to other groups. However, the mortality rate associated with this treatment regimen was low. In conclusion, the present data provide insufficient evidence regarding the beneficial effects of using HIPEC following CRS treatment. Therefore, further studies are required to determine the benefits of HIPEC for CPC patients.
Collapse
Affiliation(s)
- Sarah S Alhumaidan
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| | - Abeer M Alharbi
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| | - Ayesha Farhana Syeda
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| | - Fatimah A Alghaidani
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| | - Manal M Almutairi
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| | - Nourah A Alharbi
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| | - Reham K Alenezi
- Faculty of Pharmacy, Unaizah College of Pharmacy, Qassim University, Unaizah, SAU
| |
Collapse
|
19
|
Song XQ, Liu ZX, Kong QY, He ZH, Zhang S. Nomogram for prediction of peritoneal metastasis risk in colorectal cancer. Front Oncol 2022; 12:928894. [PMID: 36419892 PMCID: PMC9676355 DOI: 10.3389/fonc.2022.928894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/24/2022] [Indexed: 09/09/2023] Open
Abstract
OBJECTIVE Peritoneal metastasis is difficult to diagnose using traditional imaging techniques. The main aim of the current study was to develop and validate a nomogram for effectively predicting the risk of peritoneal metastasis in colorectal cancer (PMCC). METHODS A retrospective case-control study was conducted using clinical data from 1284 patients with colorectal cancer who underwent surgery at the First Affiliated Hospital of Guangxi Medical University from January 2010 to December 2015. Least absolute shrinkage and selection operator (LASSO) regression was applied to optimize feature selection of the PMCC risk prediction model and multivariate logistic regression analysis conducted to determine independent risk factors. Using the combined features selected in the LASSO regression model, we constructed a nomogram model and evaluated its predictive value via receiver operating characteristic (ROC) curve analysis. The bootstrap method was employed for repeated sampling for internal verification and the discrimination ability of the prediction models evaluated based on the C-index. The consistency between the predicted and actual results was assessed with the aid of calibration curves. RESULTS Overall, 96 cases of PMCC were confirmed via postoperative pathological diagnosis. Logistic regression analysis showed that age, tumor location, perimeter ratio, tumor size, pathological type, tumor invasion depth, CEA level, and gross tumor type were independent risk factors for PMCC. A nomogram composed of these eight factors was subsequently constructed. The calibration curve revealed good consistency between the predicted and actual probability, with a C-index of 0.882. The area under the curve (AUC) of the nomogram prediction model was 0.882 and its 95% confidence interval (CI) was 0.845-0.919. Internal validation yielded a C-index of 0.868. CONCLUSION We have successfully constructed a highly sensitive nomogram that should facilitate early diagnosis of PMCC, providing a robust platform for further optimization of clinical management strategies.
Collapse
Affiliation(s)
- Xian-qing Song
- General Surgery Department, Ningbo Fourth Hospital, Ningbo, Zhejiang, China
| | - Zhi-xian Liu
- Proctology Department, Beilun People’s Hospital of Ningbo, Ningbo, Zhejiang, China
| | - Qing-yuan Kong
- General Surgery Department, Baoan People’s Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Zhen-hua He
- General Surgery Department, Hezhou People’s Hospital, Hezhou, Guangxi, China
| | - Sen Zhang
- Department of Colorectal Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
20
|
Zwanenburg ES, Gehrels AM, Bastiaenen VP, Aalbers AGJ, Arjona-Sánchez A, Bellato V, van der Bilt JDW, D'Hoore AD, Espinosa-Redondo E, Klaver CEL, Kusters M, Nagtegaal ID, van Ramshorst B, van Santvoort HC, Sica GS, Snaebjornsson P, Wasmann KATGM, de Wilt JHW, Wolthuis AM, Tanis PJ. Metachronous peritoneal metastases in patients with pT4b colon cancer: An international multicenter analysis of intraperitoneal versus retroperitoneal tumor invasion. Eur J Surg Oncol 2022; 48:2023-2031. [PMID: 35729015 DOI: 10.1016/j.ejso.2022.05.028] [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: 04/14/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND It was hypothesized that colon cancer with only retroperitoneal invasion is associated with a low risk of peritoneal dissemination. This study aimed to compare the risk of metachronous peritoneal metastases (mPM) between intraperitoneal and retroperitoneal invasion. METHODS In this international, multicenter cohort study, patients with pT4bN0-2M0 colon cancer who underwent curative surgery were categorized as having intraperitoneal invasion (e.g. bladder, small bowel, stomach, omentum, liver, abdominal wall) or retroperitoneal invasion only (e.g. ureter, pancreas, psoas muscle, Gerota's fascia). Primary outcome was 5-year mPM cumulative rate, assessed by Kaplan-Meier analysis. RESULTS Out of 907 patients with pT4N0-2M0 colon cancer, 198 had a documented pT4b category, comprising 170 patients with intraperitoneal invasion only, 12 with combined intra- and retroperitoneal invasion, and 16 patients with retroperitoneal invasion only. At baseline, only R1 resection rate significantly differed: 4/16 for retroperitoneal invasion only versus 8/172 for intra- +/- retroperitoneal invasion (p = 0.010). Overall, 22 patients developed mPM during a median follow-up of 45 months. Two patients with only retroperitoneal invasion developed mPM, both following R1 resection. The overall 5-year mPM cumulative rate was 13% for any intraperitoneal invasion and 14% for retroperitoneal invasion only (Log Rank, p = 0.878), which was 13% and 0%, respectively, in patients who had an R0 resection (Log Rank, p = 0.235). CONCLUSION This study suggests that pT4b colon cancer patients with only retroperitoneal invasion who undergo an R0 resection have a negligible risk of mPM, but this is difficult to prove because of its rarity. This observation might have implications regarding individualized follow-up.
Collapse
Affiliation(s)
- E S Zwanenburg
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - A M Gehrels
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - V P Bastiaenen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - A G J Aalbers
- Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Arjona-Sánchez
- Unit of Surgical Oncology, Department of Surgery, Reina Sofia University Hospital and GE09 Research in Peritoneal and Retroperitoneal Oncological Surgery, (IMIBIC), Cordoba, Spain
| | - V Bellato
- Department of Surgical Science, University Hospital Tor Vergata, Rome, Italy
| | - J D W van der Bilt
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Surgery, Flevoziekenhuis, Almere, the Netherlands
| | - A D D'Hoore
- Department of Abdominal Surgery, University Hospital Leuven, Leuven, Belgium
| | - E Espinosa-Redondo
- Unit of Surgical Oncology, Department of Surgery, Reina Sofia University Hospital and GE09 Research in Peritoneal and Retroperitoneal Oncological Surgery, (IMIBIC), Cordoba, Spain
| | - C E L Klaver
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - M Kusters
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - I D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - B van Ramshorst
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - H C van Santvoort
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - G S Sica
- Department of Surgical Science, University Hospital Tor Vergata, Rome, Italy
| | - P Snaebjornsson
- Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - K A T G M Wasmann
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - J H W de Wilt
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A M Wolthuis
- Department of Abdominal Surgery, University Hospital Leuven, Leuven, Belgium
| | - P J Tanis
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Oncological and Gastrointestinal Surgery, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
21
|
Management of Peritoneal Disease in Colorectal Cancer. Hematol Oncol Clin North Am 2022; 36:569-582. [DOI: 10.1016/j.hoc.2022.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Karlsson S, Nyström H. The extracellular matrix in colorectal cancer and its metastatic settling – alterations and biological implications. Crit Rev Oncol Hematol 2022; 175:103712. [DOI: 10.1016/j.critrevonc.2022.103712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
|
23
|
Benoist H, Eveno C, Wilson S, Vigneron N, Guilloit JM, Morello R, Simon N, Odou P, Saint-Lorant G. Perception, knowledge and protective practices for surgical staff handling antineoplastic drugs during HIPEC and PIPAC. Pleura Peritoneum 2022; 7:77-86. [PMID: 35812009 PMCID: PMC9166181 DOI: 10.1515/pp-2021-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/13/2022] [Indexed: 11/28/2022] Open
Abstract
Objectives Two surgical techniques used for peritoneal metastasis involve a risk of exposure to antineoplastic drugs (ADs): hyperthermic intraperitoneal chemotherapy (HIPEC) and pressurized intraperitoneal aerosol chemotherapy (PIPAC). The objective of this study was to assess the differences in perception, training, and knowledge of the risks as well as in the protection practices and occupational exposures of all worker categories. Methods This descriptive study, led in two hospitals from two distant French regions, was performed through a face-to-face interview and assessed the perception, knowledge and handling practices of ADs by a questionnaire consisting of 52 questions. Results Fifty-one professionals participated in this survey. A total of 29.4% (n=15) professionals were afraid to handle ADs. Very few workers have been trained on handling ADs during initial training dedicated to all caregiver (5.9%; n=3). HIPEC is considered to involve a higher risk of exposure to ADs than PIPAC (81.6% (n=31) vs. 57.9% (n=22), respectively, p=0.022, agreement 65.8%). Protective equipment is considered to be less suitable for HIPEC than for PIPAC (29% (n=11) vs. 10.5% (n=4), respectively, p=0.016, agreement 81.6%). Concerning the potential AD contamination location, the participants identified a significant difference between these two practices. During HIPEC, 15.7% (n=6) of caregivers indicated that they had negative symptoms perceived in their practice vs. 2.6% (n=1) during PIPAC. Conclusions This study shows that perception, knowledge and protection practices are different between HIPEC and PIPAC. It also shows a difference between the worker categories. In view of the difficulties in making operating room staff available, the related training programmes must have an adapted format.
Collapse
Affiliation(s)
- Hubert Benoist
- Normandie Université, UNICAEN, UNIROUEN, ABTE, Centre de Lutte contre le Cancer F. Baclesse , Caen , France
| | | | - Sarah Wilson
- CHU de Caen Normandie, Normandie Université, UNICAEN, Biostatistic and Clinical Research , Caen , France
| | - Nicolas Vigneron
- CHU de Caen Normandie, Normandie Université, UNICAEN, Biostatistic and Clinical Research , Caen , France
| | - Jean-Marc Guilloit
- Department of Surgery , Comprehensive Cancer Center F. Baclesse , Caen , France
| | - Rémy Morello
- CHU de Caen Normandie, Normandie Université, UNICAEN, Biostatistic and Clinical Research , Caen , France
| | - Nicolas Simon
- Univ. Lille, CHU Lille, ULR 7365 – GRITA – Groupe de Recherche sur les Formes Injectables et les Technologies Associées , Lille , France
| | - Pascal Odou
- Univ. Lille, CHU Lille, ULR 7365 – GRITA – Groupe de Recherche sur les Formes Injectables et les Technologies Associées , Lille , France
| | - Guillaume Saint-Lorant
- Normandie Université, UNICAEN, UNIROUEN, ABTE, Centre de Lutte contre le Cancer F. Baclesse , Caen , France
- Univ. Lille, CHU Lille, ULR 7365 – GRITA – Groupe de Recherche sur les Formes Injectables et les Technologies Associées , Lille , France
| |
Collapse
|
24
|
Ravn S, Thaysen HV, Verwaal VJ, Seibæk L, Iversen LH. Cancer follow-up supported by patient-reported outcomes in patients undergoing intended curative complex surgery for advanced cancer. J Patient Rep Outcomes 2021; 5:120. [PMID: 34748095 PMCID: PMC8575728 DOI: 10.1186/s41687-021-00391-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/17/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND AIM Patient activation (PA) and Patient Involvement (PI) are considered elements in good survivorship. We aimed to evaluate the effect of a follow-up supported by electronic patient-reported outcomes (ePRO) on PA and PI. METHOD From February 2017 to January 2019, we conducted an explorative interventional study. We included 187 patients followed after intended curative complex surgery for advanced cancer at two different Departments at a University Hospital. Prior to each follow-up consultation, patients used the ePRO to screen themselves for clinical important symptoms, function and needs. The ePRO was graphically presented to the clinician during the follow-up, aiming to facilitate patient activation and involvement in each follow-up. PA was measured by the Patient Activation Measurement (PAM), while PI was measured by five indicator questions. PAM and PI data compared between (- ePRO) and interventional (+ ePRO) consultations. PAM data were analysed using a linear mixed effect regression model with intervention (yes/no) and time along with the interaction between them as categorical fixed effects. The analyses were further adjusted for time (days) since surgery. RESULTS According to our data, ePRO supported consultations did not improve PA. The average mean difference in PAM score between + ePRO and - ePRO consultations were - 0.2 (95% confidence interval - 2.6; 2.2, p = 0.9). There was no statistically significant improvement in PAM scores over time in neither + ePRO nor - ePRO group (p = 0.5). Based on the five PI-indicator questions, the majority of all consultations were evaluated as "some, much or very much" involved in consultation; providing a wider scope of dialogue, encouraged patients to ask questions and share their experiences and concerns. Nevertheless, another few patients reported not to be involved at all in the consultations. CONCLUSION We did not demonstrate evidence for ePRO supported consultations to improve patient activation, and patient activation did not improve over time. Our results generate the hypotheses that factors related to ePRO supported consultation had the potential to support PI by offering a wider scope of dialogue, and encourage patients to ask questions and share their experiences and concerns during follow-up.
Collapse
Affiliation(s)
- Sissel Ravn
- Department of Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Henriette Vind Thaysen
- Department of Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Victor Jilbert Verwaal
- Department of Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | | | - Lene Seibæk
- Department of Gynaecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | - Lene Hjerrild Iversen
- Department of Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
25
|
Lee YS, Lee WS, Kim CW, Lee SJ, Yang H, Kong SJ, Ning J, Yang KM, Kang B, Kim WR, Chon HJ, Kim C. Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer. J Immunother Cancer 2021; 8:jitc-2020-000857. [PMID: 33199510 PMCID: PMC7670945 DOI: 10.1136/jitc-2020-000857] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/14/2022] Open
Abstract
Background Peritoneal carcinomatosis (PC) is a common and devastating manifestation of colon cancer and refractory to conventional anticancer therapeutics. During the peritoneal dissemination of colon cancer, peritoneal immunity is nullified by various mechanisms of immune evasion. Here, we employed the armed oncolytic vaccinia virus mJX-594 (JX) to rejuvenate the peritoneal antitumor immune responses in the treatment of PC. Methods PC model of MC38 colon cancer was generated and intraperitoneally treated with JX and/or anti-programmed cell death protein 1 (PD-1) antibody. The peritoneal tumor burden, vascular leakage, and malignant ascites formation were then assessed. Tumors and peritoneal lavage cells were analyzed by flow cytometry, multiplex tissue imaging, and a NanoString assay. Results JX treatment effectively suppressed peritoneal cancer progression and malignant ascites formation. It also restored the peritoneal anticancer immunity by activating peritoneal dendritic cells (DCs) and CD8+ T cells. Moreover, JX selectively infected and killed peritoneal colon cancer cells and promoted the intratumoral infiltration of DCs and CD8+ T cells into peritoneal tumor nodules. JX reinvigorates anticancer immunity by reprogramming immune-related transcriptional signatures within the tumor microenvironment. Notably, JX cooperates with immune checkpoint inhibitors (ICIs), anti-programmed death-1, anti-programmed death-ligand 1, and anti-lymphocyte-activation gene-3 to elicit a stronger anticancer immunity that eliminates peritoneal metastases and malignant ascites of colon cancer compared with JX or ICI alone. Conclusions Intraperitoneal immunotherapy with JX restores peritoneal anticancer immunity and potentiates immune checkpoint blockade to suppress PC and malignant ascites in colon cancer.
Collapse
Affiliation(s)
- Yu Seong Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea (the Republic of)
| | - Won Suk Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Chang Woo Kim
- Kyung Hee University Gangdong Hospital, Gangdong-gu, Korea (the Republic of)
| | - Seung Joon Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea (the Republic of)
| | - Hannah Yang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - So Jung Kong
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - John Ning
- SillaJen Biotherapeutics, San Francisco, California, USA
| | | | - Beodeul Kang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Woo Ram Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| |
Collapse
|
26
|
Gajardo JA, Matute J, Charles R, Devaud N, Hoefler S, Schmied S, Rigo-Righi D, Butte JM. Starting a peritoneal carcinomatosis treatment program in a developing country: A prospective analysis. J Surg Oncol 2021; 124:1154-1160. [PMID: 34324203 DOI: 10.1002/jso.26626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/31/2021] [Accepted: 07/18/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) has become a valuable treatment strategy for selected patients with peritoneal carcinomatosis (PC). In Chile, it is an emerging technique. The aim of this study is to describe our protocol and report our perioperative results. METHODS A prospectively maintained database for patients undergoing exploratory surgery for PC was reviewed. Eligible patients were selected using the peritoneal cancer index in correlation with the primary tumor. Patients underwent HIPEC using mitomycin C. Clinical data and postoperative results were analyzed. RESULTS Seventy-six patients underwent exploratory surgery. Most patients were female (55%) with a median age of 62 years (range, 25-83). Complete CRS and HIPEC were achieved in 53 patients. The most frequent primary tumor site was colon-rectum (49%). The median number of resected organs was 4 (range, 1-13). Overall 90-day incidence of major complications was 26%. After a median follow-up of 26 months, 44 patients (83%) in the resected group were alive with no evidence of disease. CONCLUSIONS The PC treatment program at our institution has been established in a safe manner, with acceptable morbidity comparable to high-volume centers. A comprehensive preoperative evaluation, careful patient selection, and a cohesive team are necessary for successful results.
Collapse
Affiliation(s)
- Jorge A Gajardo
- Gastrointestinal Surgical Oncology Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Jorge Matute
- Gastrointestinal Surgical Oncology Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Roberto Charles
- Gastrointestinal Surgical Oncology Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Nicolás Devaud
- Gastrointestinal Surgical Oncology Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Sebastian Hoefler
- Gastrointestinal Surgical Oncology Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Silvia Schmied
- Anesthesia Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Daniella Rigo-Righi
- Anesthesia Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| | - Jean M Butte
- Gastrointestinal Surgical Oncology Service, Instituto Oncológico Fundación Arturo López Pérez, Santiago, Chile
| |
Collapse
|
27
|
Lurvink RJ, Rijken A, Bakkers C, Aarts MJ, Kunst PWA, van de Borne BE, van Erning FN, de Hingh IHJT. Synchronous peritoneal metastases from lung cancer: incidence, associated factors, treatment and survival: a Dutch population-based study. Clin Exp Metastasis 2021; 38:295-303. [PMID: 33738641 PMCID: PMC8179897 DOI: 10.1007/s10585-021-10085-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/03/2021] [Indexed: 01/07/2023]
Abstract
Peritoneal metastases (PM) from lung cancer are rare and it is unknown how they affect the prognosis of patients with lung cancer. This population-based study aimed to assess the incidence, associated factors, treatment and prognosis of PM from lung cancer. Data from the Netherlands Cancer Registry were used. All patients diagnosed with lung cancer between 2008 and 2018 were included. Logistic regression analysis was performed to identify factors associated with the presence of PM. Cox regression analysis was performed to identify factors associated with the overall survival (OS) of patients with PM. Between 2008 and 2018, 129,651 patients were diagnosed with lung cancer, of whom 2533 (2.0%) patients were diagnosed with PM. The European Standardized Rate of PM increased significantly from 0.6 in 2008 to 1.4 in 2018 (p < 0.001). Age between 50 and 74 years, T3-4 tumour stage, N2-3 nodal stage, tumour morphology of a small cell lung cancer or adenocarcinoma, and the presence of systemic metastases were associated with the presence of PM. The median OS of patients with PM was 2.5 months. Older age, male sex, T3-4 tumour stage, N2-3 nodal stage, not receiving systemic treatment, and the presence of systemic metastases were associated with a worse OS. Synchronous PM were diagnosed in 2.0% of patients with lung cancer and resulted in a very poor survival.
Collapse
Affiliation(s)
- Robin J Lurvink
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Anouk Rijken
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, The Netherlands
| | - Checca Bakkers
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, The Netherlands
| | - Mieke J Aarts
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Peter W A Kunst
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
- Department of Respiratory Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Ben E van de Borne
- Department of Pulmonology, Catharina Hospital, Eindhoven, The Netherlands
| | - Felice N van Erning
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, PO Box 1350, 5602 ZA, Eindhoven, The Netherlands.
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands.
- GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
28
|
Goldberg EM, Berger Y, Sood D, Kurnit KC, Kim JS, Lee NK, Yamada SD, Turaga KK, Eng OS. Differences in Sociodemographic Disparities Between Patients Undergoing Surgery for Advanced Colorectal or Ovarian Cancer. Ann Surg Oncol 2021; 28:7795-7806. [PMID: 33959831 DOI: 10.1245/s10434-021-10086-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/16/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Cytoreductive surgery (CRS) for ovarian cancer with peritoneal metastases (OPM) is an established treatment, yet access-related racial and socioeconomic disparities are well documented. CRS for colorectal cancer with peritoneal metastases (CRPM) is garnering more widespread acceptance, and it is unknown what disparities exist with regards to access. METHODS This retrospective cross-sectional multicenter study analyzed medical records from the National Cancer Database from 2010 to 2015. Patients diagnosed with CRPM or ORP only and either no or confirmed resection were included. Patient- and facility-level characteristics were analyzed using uni- and multivariable logistic regressions to identify associations with receipt of CRS. RESULTS A total of 6634 patients diagnosed with CRPM and 14,474 diagnosed with OPM were included in this study. Among patients with CRPM, 18.1% underwent CRS. On multivariable analysis, female gender (odds ratio [95% CI] 2.04 [1.77-2.35]; P < 0.001) and treatment at an academic or research facility (OR 1.55 [1.17-2.05]; P = 0.002) were associated with CRS. Among patients with OPM, 87.1% underwent CRS. On multivariable analysis, treatment at facilities with higher-income patient populations was positively associated with CRS, while age (OR 0.97 [0.96-0.98]; P < .0001), use of nonprivate insurance (OR 0.69 [0.56-0.85]; P = 0.001), and listed as Black (OR 0.62 [0.45-0.86]; P = 0.004) were negatively associated with CRS. CONCLUSION There were more systemic barriers to CRS for patients with OPM than for patients with CRPM. As CRS becomes more widely practiced for CRPM, it is likely that more socioeconomic and demographic barriers will be elucidated.
Collapse
Affiliation(s)
- Ellen M Goldberg
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Yaniv Berger
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Divya Sood
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Katherine C Kurnit
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Josephine S Kim
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Nita K Lee
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - S Diane Yamada
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Kiran K Turaga
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Oliver S Eng
- Department of Surgery, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
29
|
Bakkers C, Simkens GAAM, De Hingh IHJT. Systemic therapy in addition to cytoreduction and hyperthermic intraperitoneal chemotherapy for colorectal peritoneal metastases: recent insights from clinical studies and translational research. J Gastrointest Oncol 2021; 12:S206-S213. [PMID: 33968438 PMCID: PMC8100702 DOI: 10.21037/jgo-20-133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
There is a lack of randomized or high-quality intention-to-treat cohort studies addressing the role of systemic therapy in addition to cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) as part of the treatment of colorectal peritoneal metastases (PM). Therefore, the choice whether or not to treat patients with systemic therapy is currently mainly based on expert opinion. As a result, treatment with neoadjuvant and/or adjuvant systemic therapy is implemented in various ways around the world. The aim of this review was to provide an overview of recent insights with regard to the systemic treatment of PM of colorectal origin obtained from clinical studies and translational research.
Collapse
Affiliation(s)
- Checca Bakkers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | | | - Ignace H. J. T. De Hingh
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
- GROW - School for Oncology and Development Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
30
|
Roy P, Mignet N, Pocard M, Boudy V. Drug delivery systems to prevent peritoneal metastasis after surgery of digestives or ovarian carcinoma: A review. Int J Pharm 2021; 592:120041. [DOI: 10.1016/j.ijpharm.2020.120041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 01/08/2023]
|
31
|
Long M, Wang W, Sun Q. A high-fat diet: an unexpected role in preventing the metastatic seeding of colorectal cancer. Signal Transduct Target Ther 2020; 5:257. [PMID: 33144573 PMCID: PMC7609628 DOI: 10.1038/s41392-020-00386-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 02/05/2023] Open
Affiliation(s)
- Min Long
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Wenjing Wang
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Qiu Sun
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China.
| |
Collapse
|
32
|
Deng L, Guo S, Li H, You X, Song Y, Su H. CA125, CEA, CA19-9, and Heteroploid Cells in Ascites Fluid May Help Diagnose Peritoneal Carcinomatosis in Patients with Gastrointestinal and Ovarian Malignancies. Cancer Manag Res 2020; 12:10479-10489. [PMID: 33122947 PMCID: PMC7588672 DOI: 10.2147/cmar.s271596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/24/2020] [Indexed: 12/27/2022] Open
Abstract
Background This study explored the value of ascites and serum CA125, CEA, and CA19-9 levels and ascites DNA ploidy analysis for the diagnosis of peritoneal carcinomatosis (PC) in patients with gastrointestinal and ovarian malignancies, which can cause ascites and may disseminate peritoneally. Methods We measured ascites and serum levels of CA125, CEA, CA19-9 and performed an ascites DNA ploidy analysis in 58 patients with PC and 44 patients without PC. Results We found that a high expression level of CA125 in ascites fluid was associated with the occurrence of PC in patients with gastrointestinal and ovarian malignancies (P<0.001), and that high CEA and CA19- 9 levels in ascites fluid were associated with PC in patients with gastrointestinal malignancies (P=0.001, P=0.002). But, these tumor marker expression levels in ascites fluid were not significantly associated with the PC stage (P>0.05). We found similar serum levels of CA125, CEA, and CA19-9 between patients with gastrointestinal and ovarian malignancies and PC and those without PC (P>0.05). We found that the presence of three or more cells with heteroploid in the ascites samples was significantly associated with PC in gastrointestinal and ovarian malignancies (P<0.001). In addition, the best ROC curves and highest AUCs were achieved by combining the CA125 level and heteroploid cell analysis results (AUC for gastrointestinal and ovarian malignancies, 0.815, AUC for gastrointestinal malignancies, 0.873). Moreover, the combined ascites CA125 level and result of heteroploid cell analysis provided the best diagnostic sensitivity and specificity for PC (75.9% and 79.5%, respectively, in gastrointestinal and ovarian malignancies; 85.0% and 86.7%, respectively, in gastrointestinal malignancies). Conclusion Ascites levels of CA125, CEA, CA19-9, and heteroploid cells can be considered valuable markers for the diagnosis of PC in patients with gastrointestinal and ovarian cancer.
Collapse
Affiliation(s)
- Lin Deng
- Department of Oncology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Shikong Guo
- Department of Orthopedics, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Hong Li
- Department of Oncology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Xianghui You
- Department of Oncology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
33
|
Fiorentini G, Sarti D, Patriti A, Eugeni E, Guerra F, Masedu F, Mackay AR, Guadagni S. Immune response activation following hyperthermic intraperitoneal chemotherapy for peritoneal metastases: A pilot study. World J Clin Oncol 2020; 11:397-404. [PMID: 32874953 PMCID: PMC7450817 DOI: 10.5306/wjco.v11.i6.397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/13/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hyperthermic intraperitoneal chemotherapy (HIPEC) for peritoneal metastases (PM) is considered to be feasible, safe and to improve survival.
AIM To investigate whether an immune response is activated following HIPEC for PM.
METHODS Six patients were enrolled in this study. Peripheral blood samples were obtained from each patient prior to (day 0) and post-procedure (day 30), and used to evaluate the number of CD3+ total, CD3+/CD4+ T-Helper, CD3+/CD8+ cytotoxic T, CD3+/CD56+ natural killer and CD19+ B lymphocyte numbers, and CD4+: CD8+ T lymphocyte ratios.
RESULTS The total numbers of CD3+, CD3+/CD4+ T-Helper, CD3+/CD8+ cytotoxic T, CD3+/CD56+ natural killer and CD19+ B lymphocytes, and CD4+: CD8+ lymphocyte ratios were increased in all but one patient 30 d following the cytoreductive surgery-HIPEC procedure, and these increases were significant (P ≤ 0.05) for CD3+/CD4+ T Helper and CD3+/CD8+ cytotoxic T lymphocyte numbers.
CONCLUSION This report provides the first evidence that HIPEC exhibits immunomodulating activity in PM patients, resulting in generalized activation of the adaptive immune response. Moreover, the majority of lymphocyte populations increased following HIPEC and continued to be elevated several weeks following the procedure, consistent with a potential authentic immunomodulating effect rather than a normal inflammatory response, to be fully characterised in future studies.
Collapse
Affiliation(s)
- Giammaria Fiorentini
- Department of Onco-Hematology, Azienda Ospedaliera “Ospedali Riuniti Marche Nord”, Pesaro 61122, Italy
| | - Donatella Sarti
- Department of Onco-Hematology, Azienda Ospedaliera “Ospedali Riuniti Marche Nord”, Pesaro 61122, Italy
| | - Alberto Patriti
- Department of General Surgery, Azienda Ospedaliera “Ospedali Riuniti Marche Nord”, Pesaro 61122, Italy
| | - Emilio Eugeni
- Department of General Surgery, Azienda Ospedaliera “Ospedali Riuniti Marche Nord”, Pesaro 61122, Italy
| | - Francesco Guerra
- Department of General Surgery, Azienda Ospedaliera “Ospedali Riuniti Marche Nord”, Pesaro 61122, Italy
| | - Francesco Masedu
- Department of Applied Clinical Sciences and Biotechnology, University of L’Aquila, L’Aquila 67100, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical Sciences and Biotechnology, University of L’Aquila, L’Aquila 67100, Italy
| | - Stefano Guadagni
- Department of Applied Clinical Sciences and Biotechnology, University of L’Aquila, L’Aquila 67100, Italy
| |
Collapse
|
34
|
Ellebæk SB, Graversen M, Detlefsen S, Lundell L, Fristrup CW, Pfeiffer P, Mortensen MB. Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC)-directed treatment of peritoneal metastasis in end-stage colo-rectal cancer patients. Pleura Peritoneum 2020; 5:20200109. [PMID: 32566727 PMCID: PMC7292236 DOI: 10.1515/pp-2020-0109] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023] Open
Abstract
Background Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC) represents a novel approach to intraperitoneal chemotherapy. Hereby results, obtained with PIPAC in patients with advanced peritoneal metastasis (PM) from colorectal cancer (CRC), are presented. Methods Data from CRC patients (n = 24) included in the prospective PIPAC-OPC1 and PIPAC-OPC2 trials are reported. Oxaliplatin 92 mg/m2 was administered at 4-6-week intervals. A CE certified nebulizer was used to aerosolize the chemotherapeutics. Outcome criteria were objective tumor response, survival and adverse events. Results Retrospective analysis of 74 PIPAC procedures carried out in 24 consecutive patients with PM from CRC included from October 2015 to February 2019. Five patients had still the primary tumor in situ, and 22 patients had received palliative systemic chemotherapy. Nineteen patients completed more than two PIPAC procedures, and objective tumor response according to the histological Peritoneal Regression Grading Score (PRGS) was observed in 67% of the patients, while 21% had stable disease. Four patients (21%) had complete response (mean PRGS = 1 and negative cytology). We recorded a median survival of 37.6 (range 7.3–48.9) months from the time of PM diagnosis, whereas it was 20.5 (range 0.13–34.7) months following the first PIPAC session. Minor postoperative complications were noted, and few were considered causally related to the PIPAC treatment. However, two cases of severe postoperative complications were recorded (urosepsis and iatrogenic bowel perforation). Conclusions PIPAC with low-dose oxaliplatin can induce objective tumor regression in selected patients with advanced PM from colorectal cancer.
Collapse
Affiliation(s)
| | - Martin Graversen
- Surgical department, Odense Universitetshospital, Odense Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense Universitetshospital, Odense, Denmark
| | - Lars Lundell
- Division of Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Claus W Fristrup
- Surgical department, Odense Universitetshospital, Odense Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense Universitetshospital, Odense, Denmark
| | - Michael B Mortensen
- Upper GI and HPB Section, Department of Surgery, Odense Universitetshospital, Odense, Denmark
| |
Collapse
|
35
|
Bakkers C, van Erning FN, Rovers KP, Nienhuijs SW, Burger JW, Lemmens VE, Aalbers AG, Kok NF, Boerma D, Brandt AR, Hemmer PH, van Grevenstein WM, de Reuver PR, Tanis PJ, Tuynman JB, de Hingh IH. Long-term survival after hyperthermic intraperitoneal chemotherapy using mitomycin C or oxaliplatin in colorectal cancer patients with synchronous peritoneal metastases: A nationwide comparative study. Eur J Surg Oncol 2020; 46:1902-1907. [PMID: 32340819 DOI: 10.1016/j.ejso.2020.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/12/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES In the Netherlands, limited variability exists in performance of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) among centers treating colorectal peritoneal metastases (PM), except for the intraperitoneal drug administration. This offers a unique opportunity to investigate any disparities in survival between the two most frequently used HIPEC regimens worldwide: mitomycin C (MMC) and oxaliplatin. METHODS This was a comparative, population-based cohort study of all Dutch patients diagnosed with synchronous colorectal PM who underwent CRS-HIPEC between 2014 and 2017. They were retrieved from the Netherlands Cancer Registry. Main outcome was overall survival (OS). The effect of the intraperitoneal drug on OS was investigated using multivariable Cox regression analysis. RESULTS In total, 297 patients treated between 2014 and 2017 were included. Among them, 177 (59.6%) received MMC and 120 (40.4%) received oxaliplatin. Only primary tumor location was different between the two groups: more left-sided colon in the Oxaliplatin group (47.5% vs. 33.3%, respectively, p=0.048). The 1-, 2- and 3-year OS were 84.6% vs. 85.8%, 61.6% vs. 63.9% and 44.7% vs. 53.5% in patients treated with MMC and oxaliplatin, respectively. Median OS was 30.7 months in the MMC group vs. 46.6 months in the oxaliplatin group (p=0.181). In multivariable analysis, no influence of intraperitoneal drug on survival was observed (adjusted HR 0.77 [0.53-1.13]). CONCLUSIONS Long-term survival between patients treated with either MMC or oxaliplatin during CRS-HIPEC was not significantly different.
Collapse
Affiliation(s)
- C Bakkers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands.
| | - F N van Erning
- Department of Research, Netherlands Comprehensive Cancer Organization, Utrecht, the Netherlands
| | - K P Rovers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - S W Nienhuijs
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - J W Burger
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - V E Lemmens
- Department of Research, Netherlands Comprehensive Cancer Organization, Utrecht, the Netherlands
| | - A G Aalbers
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - N F Kok
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - A R Brandt
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - P H Hemmer
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - W M van Grevenstein
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - P R de Reuver
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P J Tanis
- Department of Surgery, Amsterdam University Medical Centers, University of Amsterdam, Cancer Centre Amsterdam, Amsterdam, the Netherlands
| | - J B Tuynman
- Department of Surgery, Amsterdam University Medical Centers, VU Medical Center, Amsterdam, the Netherlands
| | - I H de Hingh
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands; GROW - School for Oncology and Development Biology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
36
|
Demuytere J, Ceelen W, Van Dorpe J, Hoorens A. The role of the peritoneal microenvironment in the pathogenesis of colorectal peritoneal carcinomatosis. Exp Mol Pathol 2020; 115:104442. [PMID: 32305340 DOI: 10.1016/j.yexmp.2020.104442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 01/06/2023]
Abstract
Recent insights have implicated mesothelial-to-mesenchymal transition (MMT) as a mechanism by which mesothelial cells can transdifferentiate into cancer-associated fibroblasts (CAFs) in several cancers metastasizing to the peritoneum. However, this was not evaluated extensively in colorectal cancer. We examined the presumed mesothelial origin of CAFs in three types of colorectal carcinoma: conventional type adenocarcinoma, mucinous carcinoma and signet ring cell carcinoma. We evaluated the expression of mesothelial, mesenchymal, angiogenesis and colorectal cancer-related markers in peritoneal samples of twelve colorectal cancer patients with peritoneal carcinomatosis and four control patients by immunohistochemistry. We observed morphological and immunohistochemical changes in the vicinity of tumor implants in all studied colorectal cancer types. Mesothelial cells acquired a spindle-shaped myofibroblast-like morphology, lost expression of mesothelial markers, and gained expression of mesenchymal markers. Analysis of consecutive tissue sections and double staining for mesothelial and mesenchymal markers revealed overlap in expression of mesothelial and CAF markers. These findings are highly suggestive of a mesothelial origin of CAFs in peritoneal carcinomatosis in colorectal cancer. Interfering with the process of MMT might be a valuable approach in treating and preventing peritoneal carcinomatosis. Differences observed between colorectal cancer types suggest that one single strategy might not be applicable.
Collapse
Affiliation(s)
- Jesse Demuytere
- Laboratory of Experimental Surgery, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Laboratory of Experimental Surgery, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium..
| |
Collapse
|
37
|
Pletcher E, Gleeson E, Labow D. Peritoneal Cancers and Hyperthermic Intraperitoneal Chemotherapy. Surg Clin North Am 2020; 100:589-613. [PMID: 32402303 DOI: 10.1016/j.suc.2020.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytoreductive surgery followed by hyperthermic intraperitoneal chemotherapy is an aggressive, potentially curative approach used to treat locoregional disease associated with primary and secondary malignancies of the peritoneum. It involves resection of all macroscopic disease larger than 2.5 mm, followed by instillation of hyperthermic chemotherapy directly into the peritoneum for higher drug exposure to microscopic disease. In select patients with primary peritoneal mesothelioma, pseudomyxoma peritonei, colorectal adenocarcinoma, appendiceal adenocarcinoma, or ovarian cancer, with no extra-abdominal metastasis and limited involvement of the peritoneum, the procedure can be performed to increase overall survival.
Collapse
Affiliation(s)
- Eric Pletcher
- Surgery Department, Mount Sinai West and Morningside, 425 West 59th Street, 7th Floor, New York, NY 10019, USA
| | - Elizabeth Gleeson
- Division of Surgical Oncology, Mount Sinai Hospital, 19 East 98th Street, Suite 7A, New York, NY 10029, USA
| | - Daniel Labow
- Surgery Department, Mount Sinai Hospital, Mount Sinai West and Morningside, 425 West 59th Street, 7th Floor, New York, NY 10019, USA.
| |
Collapse
|
38
|
Iodine overlays to improve differentiation between peritoneal carcinomatosis and benign peritoneal lesions. Eur Radiol 2020; 30:3968-3976. [DOI: 10.1007/s00330-020-06729-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022]
|
39
|
Ravn S, Heide-Jørgensen U, Christiansen CF, Verwaal VJ, Hagemann-Madsen RH, Iversen LH. Overall risk and risk factors for metachronous peritoneal metastasis after colorectal cancer surgery: a nationwide cohort study. BJS Open 2020; 4:284-292. [PMID: 32207578 PMCID: PMC7093782 DOI: 10.1002/bjs5.50247] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 11/12/2019] [Indexed: 01/16/2023] Open
Abstract
Background This study aimed to identify the cumulative incidence and risk factors of metachronous peritoneal metastasis (M‐PM) from colorectal cancer in patients who had intended curative treatment. Methods Patients with colorectal cancer were identified using the Danish Colorectal Cancer Group database for 2006–2015. The Danish Pathology Registry and the Danish National Patient Registry were used to identify M‐PM to 2017. Risk factors were estimated by multivariable absolute risk regression, treating death and other cancers as competing risks. Overall risk and risk differences (RDs) were estimated at 1, 3 and 5 years. Results In 22 586 patients with colorectal cancer, the overall risk of M‐PM was reported to be 0·9 (95 per cent c.i. 0·8 to 1·0) per cent at 1 year, 1·9 (1·8 to 2·1) per cent at 3 years and 2·2 (2·0 to 2·4) per cent at 5 years. Advanced tumour category ((y)pT4 versus (y)pT1) increased the RD of both M‐PM (2·9 (95 per cent c.i. 2·1 to 3·7) at 1 year and 6·0 (4·9 to 7·2) at 3 years) and lymph node involvement ((y)pN2 versus (y)pN0) (2·5 (1·8 to 3·2) at year and 4·3 (3·2 to 5·3) at 3 years). No further increase in risk was observed at 5 years. In a subanalysis, tumour‐involved resection margin (R1 versus R0) was associated with M‐PM with a RD of 3·9 (1·6 to 6·2) at 1 year and 5·9 (2·6 to 9·3) at 3 years. Conclusion The overall risk of M‐PM in patients with colorectal cancer is low, but is increased in advanced T and N status. Follow‐up of at least 3 years after colorectal cancer surgery may be necessary, given the potential curative treatment of early diagnosed M‐PM.
Collapse
Affiliation(s)
- S Ravn
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - U Heide-Jørgensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - C F Christiansen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - V J Verwaal
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | | | - L H Iversen
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark.,Danish Colorectal Cancer Group, Copenhagen, Denmark
| |
Collapse
|
40
|
Demontoux L, Derangère V, Pilot T, Thinselin C, Chevriaux A, Chalmin F, Bouyer F, Ghiringhelli F, Rébé C. Hypotonic stress enhances colon cancer cell death induced by platinum derivatives and immunologically improves antitumor efficacy of intraperitoneal chemotherapy. Int J Cancer 2019; 145:3101-3111. [PMID: 31344262 DOI: 10.1002/ijc.32590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 12/24/2022]
Abstract
Colorectal cancer is a highly metastatic disease that could invade various distal organs and also the peritoneal cavity leading to peritoneal carcinomatosis. This is a terminal condition with poor prognosis and only palliative treatments such as cytoreductive surgery and intraperitoneal chemotherapy are proposed to some patients. However, clinicians use different parameters of treatments without any consensus. Here we decided to evaluate the effect of osmolarity in the efficacy of this procedure to kill colon cancer cells. We first show that a short exposure of platinum derivatives in hypotonic conditions is more efficient to decrease cell viability of human and murine colon cancer cells in vitro as compared to isotonic conditions. This is related to more important incorporation of platinum and the capacity of hypotonic stress to induce the copper transporter CTR1 oligomerization. Oxaliplatin in hypotonic conditions induces caspase-dependent cell death of colon cancer cells. Moreover, hypotonic conditions also modulate the capacity of oxaliplatin and cisplatin (but not carboplatin) to induce immunogenic cell death (ICD). In vivo, oxaliplatin in hypotonic conditions increases CD8+ T cell tumor infiltration and activation. Finally, in a murine peritoneal carcinomatosis model, oxaliplatin in hypotonic conditions is the only tested protocol which is able to slow down the appearance of tumor nodules and increase mice survival, while showing no effect in CD8+ T cells depleted mice or in immunodeficient mice. Altogether, our study provides new information both in vitro and in a preclinical model of peritoneal carcinomatosis, which highlights the importance of hypoosmolarity in intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Lucie Demontoux
- INSERM LNC-UMR1231, Dijon, France.,University of Bourgogne Franche-Comté, Dijon, France
| | - Valentin Derangère
- INSERM LNC-UMR1231, Dijon, France.,University of Bourgogne Franche-Comté, Dijon, France.,Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, Dijon, France
| | - Thomas Pilot
- INSERM LNC-UMR1231, Dijon, France.,University of Bourgogne Franche-Comté, Dijon, France
| | | | - Angélique Chevriaux
- INSERM LNC-UMR1231, Dijon, France.,Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, Dijon, France
| | - Fanny Chalmin
- INSERM LNC-UMR1231, Dijon, France.,University of Bourgogne Franche-Comté, Dijon, France
| | | | - François Ghiringhelli
- INSERM LNC-UMR1231, Dijon, France.,University of Bourgogne Franche-Comté, Dijon, France.,Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, Dijon, France
| | - Cédric Rébé
- INSERM LNC-UMR1231, Dijon, France.,University of Bourgogne Franche-Comté, Dijon, France.,Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
41
|
Klos D, Riško J, Loveček M, Skalický P, Svobodová I, Krejčí D, Melichar B, Mohelníková-Duchoňová B, Lemstrová R. Trends in peritoneal surface malignancies: evidence from a Czech nationwide population-based study. World J Surg Oncol 2019; 17:182. [PMID: 31694646 PMCID: PMC6836516 DOI: 10.1186/s12957-019-1731-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023] Open
Abstract
Background The aim of this study is to identify the incidence trends of primary and secondary peritoneal surface malignancies in a representative Czech population. Methods Data were obtained from patients registered in the Czech National Cancer Registry between 1979 and 2016. The incidence rates were analyzed between 2012 and 2016. To observe the incidence trends, we analyzed the data from two time periods, 1979–2005 and 2006–2016. The analyzed data included age, sex, and the histological types and primary origins of the malignancies. The Cochrane-Armitage test for linear trends was used for verification of the null hypothesis. The significance level established for hypothesis testing was p = 0.05. Results Between 2012 and 2016, 230 patients with primary peritoneal tumors were identified and divided into the following groups according to their “International Statistical Classification of Diseases and Related Health Problems, 10th revision” codes: malignant neoplasm of specified parts of the peritoneum (C48.1); malignant neoplasm of the peritoneum, unspecified (C48.2); and malignant neoplasm of overlapping sites of the retroperitoneum and peritoneum (C48.8). Moreover, 549 primary tumors of the appendix (C18.1, encompassing all appendiceal malignancies) and 3137 secondary synchronous peritoneal carcinomatoses of other primary origins were documented. The age-adjusted incidence of primary peritoneal tumors in 2012–2016 was 4.36/year/1,000,000 inhabitants. The age-adjusted incidence of synchronous secondary peritoneal malignancies in 2014–2016 was 99.0/year/1,000,000 inhabitants. The diagnoses of primary peritoneal malignancies followed a stable trend between 1979 and 2016. However, the incidences of primary tumors of the appendix increased by 76.7%. Conclusions The data produced in our study ought to clarify the status of peritoneal surface malignancies in the Czech Republic, which can lead to improved planning and development of therapeutic interventions as well as physician training.
Collapse
Affiliation(s)
- Dušan Klos
- Department of Surgery I, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Juraj Riško
- Department of Surgery I, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Martin Loveček
- Department of Surgery I, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Pavel Skalický
- Department of Surgery I, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Ivana Svobodová
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | - Denisa Krejčí
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, CZ-779 00, Olomouc, Czech Republic
| | - Beatrice Mohelníková-Duchoňová
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, CZ-779 00, Olomouc, Czech Republic.,Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Radmila Lemstrová
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, CZ-779 00, Olomouc, Czech Republic.
| |
Collapse
|
42
|
Nanotechnology is an important strategy for combinational innovative chemo-immunotherapies against colorectal cancer. J Control Release 2019; 307:108-138. [DOI: 10.1016/j.jconrel.2019.06.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/12/2019] [Accepted: 06/16/2019] [Indexed: 12/15/2022]
|
43
|
Steffen T, Putora PM, Hübner M, Gloor B, Lehmann K, Kettelhack C, Adamina M, Peterli R, Schmidt J, Ris F, Glatzer M. Diagnostic Nodes of Patient Selection for Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy Among Colorectal Cancer Patients: A Swiss National Multicenter Survey. Clin Colorectal Cancer 2019; 18:e335-e342. [PMID: 31371166 DOI: 10.1016/j.clcc.2019.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The management of patients with colorectal cancer (CRC) with peritoneal metastases is challenging, and the roles of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are unclear and debated among experts. MATERIALS AND METHODS The experts of the Swiss Peritoneal Cancer Group were contacted and agreed to participate in this analysis. Experts from 9 centers in Switzerland provided their decision algorithms for CRS/HIPEC for patients with or at high risk for peritoneal metastases from CRC. Their responses were converted into decision trees on the basis of objective consensus methodology. The decision trees were used as a basis to identify consensus and discrepancies. RESULTS The final treatment algorithms included a total of 5 decision criteria (age, Peritoneal Cancer Index [PCI], extraperitoneal metastases, Peritoneal Surface Disease Severity Score, and various risk factors [RF]) and 2 treatment options (HIPEC, yes or no). HIPEC was never recommended for patients without peritoneal metastases in the absence of RF for peritoneal metastases. For patients with a PCI ≤15 without organ metastases, all centers recommended CRS/HIPEC. There was also a consensus not to perform CRS/HIPEC in elderly patients (80 years and older), those with a PCI >20, and those with unresectable metastases. For patients with a PCI = 16 to 20, there was no consensus. CONCLUSION Multiple decision criteria relevant to all participating centers were identified. Because patient selection for CRS/HIPEC remains difficult, uniform criteria for the term "high risk" for peritoneal metastases and systemic metastases are helpful. Future trials and guidelines should take these criteria into account.
Collapse
Affiliation(s)
- Thomas Steffen
- Department of Surgery, Kantonsspital St Gallen, St Gallen, Switzerland.
| | - Paul Martin Putora
- Department of Radiation Oncology, Kantonsspital St Gallen, St Gallen, Switzerland; Department of Radiation Oncology, University of Bern, Bern, Switzerland
| | - Martin Hübner
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Beat Gloor
- Department of Surgery, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Kuno Lehmann
- Department of Surgery and Transplantation, University Hospital of Zürich, Zürich, Switzerland
| | | | - Michel Adamina
- Department of Surgery, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Ralph Peterli
- Department of Surgery, St Claraspital, Basel, Switzerland
| | - Jan Schmidt
- Department of Surgery, Klinik Hirslanden, Zürich, Switzerland
| | - Frédéric Ris
- Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
| | - Markus Glatzer
- Department of Radiation Oncology, Kantonsspital St Gallen, St Gallen, Switzerland
| |
Collapse
|
44
|
Pinto A, Pocard M. Hyperthermic intraperitoneal chemotherapy with cisplatin and mitomycin C for colorectal cancer peritoneal metastases: A systematic review of the literature. Pleura Peritoneum 2019; 4:20190006. [PMID: 31388562 PMCID: PMC6668656 DOI: 10.1515/pp-2019-0006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/10/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The randomized trial PRODIGE 7 failed to show the benefit of oxaliplatin hyperthermic intraperitoneal chemotherapy (HIPEC) in colorectal peritoneal metastasis treatment (CR PM). This systematic review focuses on the association of cisplatin (CDDP) with mitomycin C (MMC) in HIPEC in CR PM. CONTENT Experimental studies demonstrated that hyperthermia, in addition to CDDP ± MMC treatment, gradually improved the cytotoxic effect by increasing early apoptosis, eATP interaction, intracellular CDDP concentration (by 20%) and p73 expression. Recent studies with highly selected patients reported unusual prolonged survival with a median overall survival (OS) of approximately 60 months, with a HIPEC combination of CDDP (25 mg/m2/L) plus MMC (3.3 mg/m2/L) at a temperature of 41.5-42.5 °C for 60-90 min. Major complications occurred in less than 30% of patients with limited hematological toxicity (less than 15%). In addition, in a phase 2 trial, an adjuvant HIPEC benefit was demonstrated in colorectal cancer patients with high risk for peritoneal failure (5-year OS: 81.3% vs. 70% for the HIPEC group vs. the control group, respectively, p=0.047). After a recurrence, an iterative procedure permitted similar recurrence-free disease (13 vs. 13.7 months) with an acceptable morbidity (18.7% of severe complications). SUMMARY AND OUTLOOK The combination of CDDP and MMC seems to be an interesting protocol as an alternative to high-dose and short-term oxaliplatin.
Collapse
Affiliation(s)
- Amandine Pinto
- Inserm U1275 - Carcinose Péritoine et Paris-Technologie, INSERM, Paris, France
| | - Marc Pocard
- U1275 - Carcinose Péritoine et Paris-Technologie, INSERM, Paris, France
| |
Collapse
|
45
|
Synchronous liver metastases and peritoneal carcinomatosis from colorectal cancer: different strategies for curative treatment? Langenbecks Arch Surg 2019; 404:477-488. [PMID: 31025165 DOI: 10.1007/s00423-019-01787-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/10/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Management of patients with resectable hepatic metastases (HMs) and colorectal peritoneal carcinomatosis (CRPC) is not currently standardised. OBJECTIVE The aims of this study were to evaluate the safety of cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS/HIPEC) and hepatic surgery for patients with CRPC with synchronous hepatic metastases (HM), and its impact on survival rates. METHODS A retrospective analysis was performed, including patients undergoing CRS/HIPEC for CRPC from 2007 to September 2016 in two groups, with (HM+) and without (HM-) synchronous hepatic metastases. Patients with extra-abdominal metastases were excluded. The hepatic strategy was described. Morbimortality and survival were compared between the two groups. RESULTS One hundred nine patients underwent CRS/HIPEC for CRPC with or without hepatic surgery with curative intent: 33 patients with (HM+) and 76 patients without (HM-) synchronous HM. The median follow-up was 30 months. All patients with HM (HM+) received neoadjuvant chemotherapy vs. 88.1% in the HM- group (p = 0.04) associated with monoclonal antibody in 66.6% of cases in the HM+ group vs. 57% in the HM- group (p = 0.01). In the HM+ group, two steps were implemented to treat peritoneal and hepatic metastases in 15 patients (45%). In this group, planned hepatic resection in two procedures was performed for eight patients, all presenting bilobar HM. Postoperative morbidity did not differ between the two groups. No deaths occurred. Median overall survival (OS) and recurrence-free survival (RFS) were 31 and 65 months (p = 0.188), versus 21 and 24 months (p = 0.119), respectively, in the HM+ versus HM- groups. In multivariate analysis, the peritoneal cancer index (PCI) was the only significant prognostic factor whereas synchronous HM was not a significant prognostic factor. CONCLUSION Curative surgical treatment for CRPC with synchronous HM seems to be feasible and safe, and could facilitate long survival rates, compared to patients without HM. The hepatic strategy is not standardised. However, a "two-step" surgical strategy could be proposed in order to reduce postoperative morbidity rates.
Collapse
|
46
|
Bastiaenen VP, Klaver CEL, Kok NFM, de Wilt JHW, de Hingh IHJT, Aalbers AGJ, Boerma D, Bremers AJA, Burger JWA, van Duyn EB, Evers P, van Grevenstein WMU, Hemmer PHJ, Madsen EVE, Snaebjornsson P, Tuynman JB, Wiezer MJ, Dijkgraaf MGW, van der Bilt JDW, Tanis PJ. Second and third look laparoscopy in pT4 colon cancer patients for early detection of peritoneal metastases; the COLOPEC 2 randomized multicentre trial. BMC Cancer 2019; 19:254. [PMID: 30898098 PMCID: PMC6429794 DOI: 10.1186/s12885-019-5408-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/25/2019] [Indexed: 12/24/2022] Open
Abstract
Background Approximately 20–30% of patients with pT4 colon cancer develop metachronous peritoneal metastases (PM). Due to restricted accuracy of imaging modalities and absence of early symptoms, PM are often detected at a stage in which only a quarter of patients are eligible for curative intent treatment. Preliminary findings of the COLOPEC trial (NCT02231086) revealed that PM were already detected during surgical re-exploration within two months after primary resection in 9% of patients with pT4 colon cancer. Therefore, second look diagnostic laparoscopy (DLS) to detect PM at a subclinical stage may be considered an essential component of early follow-up in these patients, although this needs confirmation in a larger patient cohort. Furthermore, a third look DLS after a negative second look DLS might be beneficial for detection of PM occurring at a later stage. Methods The aim of this study is to determine the yield of second look DLS and added value of third look DLS after negative second look DLS in detecting occult PM in pT4N0-2 M0 colon cancer patients after completion of primary treatment. Patients will undergo an abdominal CT at 6 months postoperative, followed by a second look DLS within 1 month if no PM or other metastases not amenable for local treatment are detected. Patients without PM will subsequently be randomized between routine follow-up including 18 months abdominal CT, or an experimental arm with a third look DLS provided that PM or incurable metastases are absent at the 18 months abdominal CT. Primary endpoint is the proportion of PM detected after a negative second look DLS and will be determined at 20 months postoperative. Discussion Second look DLS is supposed to result in 10% occult PM, and third look DLS after negative second look DLS is expected to detect an additional 10% of PM compared to routine follow-up alone in patients with pT4 colon cancer. Detection of PM at an early stage will likely increase the proportion of patients eligible for curative intent treatment and subsequently improve survival, given the uniformly reported direct association between the extent of peritoneal disease and survival. Trial registration ClinicalTrials.gov: NCT03413254, January 2018. Electronic supplementary material The online version of this article (10.1186/s12885-019-5408-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivian P Bastiaenen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, PO Box 22660, 1105 AZ, Amsterdam, the Netherlands.
| | - Charlotte E L Klaver
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, PO Box 22660, 1105 AZ, Amsterdam, the Netherlands
| | - Niels F M Kok
- Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | - Arend G J Aalbers
- Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Andre J A Bremers
- Department of Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | - Eino B van Duyn
- Department of Surgery, Medisch Spectrum Twente, Enschede, the Netherlands
| | - Pauline Evers
- Dutch Federation of Cancer Patient Organizations (NFK), Utrecht, the Netherlands
| | | | - Patrick H J Hemmer
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Eva V E Madsen
- Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jurriaan B Tuynman
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Marinus J Wiezer
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Marcel G W Dijkgraaf
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jarmila D W van der Bilt
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, PO Box 22660, 1105 AZ, Amsterdam, the Netherlands.,Department of Surgery, Flevo hospital, Almere, the Netherlands
| | - Pieter J Tanis
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, PO Box 22660, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
47
|
Zihui Yong Z, Ching GTH, Ching MTC. Metastatic Profile of Colorectal Cancer: Interplay Between Primary Tumor Location and KRAS Status. J Surg Res 2019; 246:325-334. [PMID: 30737098 DOI: 10.1016/j.jss.2018.11.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/06/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Mutant KRAS tumors are purported to metastasize differently than wild-type KRAS tumors. The biological heterogeneity of tumors from different parts of the colon are also reported to affect metastasis. This study aims to characterize the metastatic profile by evaluating these factors in unison. METHODS Retrospective analysis of 899 patients with metastatic colorectal cancers treated from January 2010 to December 2014 was conducted. KRAS mutation status and primary tumors location were correlated with single-site metastasis (liver, lung, and peritoneum) and dual-site metastases (liver-peritoneum, liver-lung, and lung-peritoneum). Patients without KRAS analyses were excluded. RESULTS Right-sided tumors had highest frequency of peritoneal metastasis as compared to left-sided or rectal tumors (34.7% versus 15.8% versus 8.8%, P = 0.00) regardless of KRAS status (32.6% versus 38.5%, P = 0.62). Left-sided tumors with wild-type KRAS had greater proportion of liver metastasis (78.6% versus 53.5%, P = 0.00), whereas those with mutant KRAS had greater proportion of lung metastasis (23.3% versus 8.7%, P = 0.02). Rectal tumors with wild-type KRAS tend to spread to the liver (81.4% versus 48.0%, P = 0.00) and not to the peritoneum (2.3% versus 20.0%, P = 0.01). In dual-site metastases, left-sided tumors with wild-type KRAS had more liver-peritoneal metastases (75.0% versus 29.4%, P = 0.00), whereas mutant KRAS had greater lung-liver metastases (64.7% versus 20.8%, P = 0.01). Rectal tumors had the predilection for lung-liver metastases as compared to right-sided and left-sided tumors (92.3% versus 40.0% versus 39.0%, P = 0.00) regardless of KRAS status (100% versus 75%, P = 0.12). CONCLUSIONS Our results may streamline surveillance programs based on primary tumor location and KRAS mutational status.
Collapse
|
48
|
Nowacki M, Zegarski W. The scientific report from the first pressurized intraperitoneal aerosol chemotherapy (PIPAC) procedures performed in the eastern part of Central Europe. J Int Med Res 2018; 46:3748-3758. [PMID: 29916281 PMCID: PMC6135997 DOI: 10.1177/0300060518778637] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/02/2018] [Indexed: 12/15/2022] Open
Abstract
Objective To perform a single-centre, detailed analysis of the preparations for the introduction of the first pressurized intraperitoneal aerosol chemotherapy (PIPAC) programme in the eastern part of Central Europe. Methods The study analysed the 14-month preparation period prior to the performance of the first PIPAC procedure with respect to: (i) general preparations; (ii) patient referral and qualification; (iii) the first PIPAC procedure; (iv) the 2 weeks following PIPAC programme establishment; and (v) general problematic issues that arose. Results The length of time needed to prepare our institution for the first PIPAC procedure was extremely long compared with other European Union PIPAC centres: 14 months versus a standard 3-6 months of preparation. The longest amount of time (12 months) was required to prepare the required paperwork. Conclusions A new PIPAC programme was successfully established in the eastern part of Central Europe. The length of time to implement this method was significantly longer because of lengthy bureaucratic processes. These current findings should help new centres, especially in this part of Europe, to establish a PIPAC programme more quickly.
Collapse
Affiliation(s)
- Maciej Nowacki
- Department of Surgical Oncology, Ludwik Rydygier’s
Collegium Medicum, Nicolaus Copernicus University in Torun, Bydgoszcz,
Poland
| | - Wojciech Zegarski
- Department of Surgical Oncology, Ludwik Rydygier’s
Collegium Medicum, Nicolaus Copernicus University in Torun, Bydgoszcz,
Poland
| |
Collapse
|
49
|
Benlahfid M, Traboulsi W, Sergent F, Benharouga M, Elhattabi K, Erguibi D, Karkouri M, Elattar H, Fadil A, Fahmi Y, Aboussaouira T, Alfaidy N. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) and its receptor PROKR2 are associated to human colorectal cancer progression and peritoneal carcinomatosis. Cancer Biomark 2018; 21:345-354. [PMID: 29226856 DOI: 10.3233/cbm-170499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The highest risk factor for mortality among malignant tumors is metastasis. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic factor which biological activity is mediated via two G protein-coupled receptors, prokineticin receptor1 (PROKR1) and PROKR2. Recent studies suggested that EG-VEGF expression is deregulated in multiple cancers including colorectal cancer (CRC). METHODS Using distinctive CRC and peritoneal carcinomatosis (PC) cohorts and a corresponding control cohort, we determined the circulating levels of EG-VEGF and its in situ expression, and that of its related receptors. RESULTS Circulating EG-VEGF levels were significantly increased in patients with metastatic PC compared to CRC and control patients (p< 0.05). Furthermore, according to clinicopathologic examinations, local EG-VEGF expression correlated with higher tumor and nodal stages (p< 0.001) of CRC. EG-VEGF and PROKR2 were highly expressed in colorectal primary lesions compared to positive controls. PROKR1 expression was lower and did not change in tumor specimens. Also, EG-VEGF and its receptor PROKR2 were differentially expressed in the colorectal primary lesions and in the control groups. CONCLUSION Altogether these findings suggest that EG-VEGF/receptors system might be an important actor in the CRC progression into PC and might be involved in the ability of tumor cells to invade other organs. Circulating EG-VEGF could be proposed as a prognostic marker in human CRC and its progression into PC.
Collapse
Affiliation(s)
- Mohammed Benlahfid
- Laboratory of Scientific and Clinical Researches in Cancerous Pathologies, Faculty of Medicine and Pharmacy, University Hassan II of Casablanca, Casablanca, Morocco
| | - Wael Traboulsi
- Institut National de la Santé et de la Recherche Médicale U1036, Grenoble, France.,University Grenoble-Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Frederic Sergent
- Institut National de la Santé et de la Recherche Médicale U1036, Grenoble, France.,University Grenoble-Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Mohamed Benharouga
- University Grenoble-Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France.,Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France
| | - Khalid Elhattabi
- Laboratory of Scientific and Clinical Researches in Cancerous Pathologies, Faculty of Medicine and Pharmacy, University Hassan II of Casablanca, Casablanca, Morocco.,Ibn Rochd University Hospital of Casablanca, University Hassan II of Casablanca, Casablanca, Morocco
| | - Driss Erguibi
- Laboratory of Scientific and Clinical Researches in Cancerous Pathologies, Faculty of Medicine and Pharmacy, University Hassan II of Casablanca, Casablanca, Morocco.,Ibn Rochd University Hospital of Casablanca, University Hassan II of Casablanca, Casablanca, Morocco
| | - Mehdi Karkouri
- Ibn Rochd University Hospital of Casablanca, University Hassan II of Casablanca, Casablanca, Morocco
| | - Hicham Elattar
- Laboratory of anatomopathology Moulay Driss 1er, Casablanca, Morocco
| | - Abdelaziz Fadil
- Ibn Rochd University Hospital of Casablanca, University Hassan II of Casablanca, Casablanca, Morocco
| | - Yassine Fahmi
- Ibn Rochd University Hospital of Casablanca, University Hassan II of Casablanca, Casablanca, Morocco
| | - Touria Aboussaouira
- Laboratory of Scientific and Clinical Researches in Cancerous Pathologies, Faculty of Medicine and Pharmacy, University Hassan II of Casablanca, Casablanca, Morocco.,Laboratory of Scientific and Clinical Researches in Cancerous Pathologies, Faculty of Medicine and Pharmacy, University Hassan II of Casablanca, Casablanca, Morocco
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale U1036, Grenoble, France.,University Grenoble-Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France.,Laboratory of Scientific and Clinical Researches in Cancerous Pathologies, Faculty of Medicine and Pharmacy, University Hassan II of Casablanca, Casablanca, Morocco
| |
Collapse
|
50
|
Wonder E, Simón-Gracia L, Scodeller P, Majzoub RN, Kotamraju VR, Ewert KK, Teesalu T, Safinya CR. Competition of charge-mediated and specific binding by peptide-tagged cationic liposome-DNA nanoparticles in vitro and in vivo. Biomaterials 2018; 166:52-63. [PMID: 29544111 PMCID: PMC5944340 DOI: 10.1016/j.biomaterials.2018.02.052] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/14/2018] [Accepted: 02/27/2018] [Indexed: 12/31/2022]
Abstract
Cationic liposome-nucleic acid (CL-NA) complexes, which form spontaneously, are a highly modular gene delivery system. These complexes can be sterically stabilized via PEGylation [PEG: poly (ethylene glycol)] into nanoparticles (NPs) and targeted to specific tissues and cell types via the conjugation of an affinity ligand. However, there are currently no guidelines on how to effectively navigate the large space of compositional parameters that modulate the specific and nonspecific binding interactions of peptide-targeted NPs with cells. Such guidelines are desirable to accelerate the optimization of formulations with novel peptides. Using PEG-lipids functionalized with a library of prototypical tumor-homing peptides, we varied the peptide density and other parameters (binding motif, peptide charge, CL/DNA charge ratio) to study their effect on the binding and uptake of the corresponding NPs. We used flow cytometry to quantitatively assess binding as well as internalization of NPs by cultured cancer cells. Surprisingly, full peptide coverage resulted in less binding and internalization than intermediate coverage, with the optimum coverage varying between cell lines. In, addition, our data revealed that great care must be taken to prevent nonspecific electrostatic interactions from interfering with the desired specific binding and internalization. Importantly, such considerations must take into account the charge of the peptide ligand as well as the membrane charge density and the CL/DNA charge ratio. To test our guidelines, we evaluated the in vivo tumor selectivity of selected NP formulations in a mouse model of peritoneally disseminated human gastric cancer. Intraperitoneally administered peptide-tagged CL-DNA NPs showed tumor binding, minimal accumulation in healthy control tissues, and preferential penetration of smaller tumor nodules, a highly clinically relevant target known to drive recurrence of the peritoneal cancer.
Collapse
Affiliation(s)
- Emily Wonder
- Materials, Physics, and Molecular, Cellular, & Developmental Biology Departments, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| | - Lorena Simón-Gracia
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia
| | - Pablo Scodeller
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ramsey N Majzoub
- Materials, Physics, and Molecular, Cellular, & Developmental Biology Departments, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| | - Venkata Ramana Kotamraju
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Kai K Ewert
- Materials, Physics, and Molecular, Cellular, & Developmental Biology Departments, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| | - Tambet Teesalu
- Materials, Physics, and Molecular, Cellular, & Developmental Biology Departments, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Cyrus R Safinya
- Materials, Physics, and Molecular, Cellular, & Developmental Biology Departments, University of California at Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|