1
|
Wu G, Standring OJ, King DA, Gholami S, Devoe CE, Thiels CA, Grotz TE, Weiss MJ, Whelan RL, Raoof M, DePeralta DK. Management of Peritoneal Metastasis in Patients with Pancreatic Ductal Adenocarcinoma. Curr Oncol 2025; 32:103. [PMID: 39996904 PMCID: PMC11854847 DOI: 10.3390/curroncol32020103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The peritoneum is the second most common site of metastasis in patients with pancreatic ductal adenocarcinoma (PDAC). Up to half of all patients that undergo curative-intent resection eventually develop peritoneal metastasis (PM), which accounts for significant morbidity and drives mortality. Despite recent advances in management, PM is associated with very poor prognosis, which is often measured in weeks to months. Clinical manifestations including bowel obstruction, ascites, and urinary obstruction have profound impact on quality of life. Even with relatively advanced disease, PM often remains occult on imaging and thus tend to be underdiagnosed and understudied. Many patients with peritoneal-only PM are excluded from clinical trials because response cannot be measured by standard radiographic criteria. Furthermore, as patients with PM are not eligible for surgical resection and low-volume peritoneal disease is often not amenable to percutaneous biopsy, tissue samples for peritoneal-specific translational studies are limited. Intraperitoneal therapeutics have been proposed as an attractive option for PM, as better penetration of tumor tissue can be achieved with less systemic toxicity compared with intravenous chemotherapy. Heated intraperitoneal chemotherapy (HIPEC), typically combined with cytoreductive surgery (CRS), is an option for select patients with PM from gynecologic or gastrointestinal primary, and for patients with primary peritoneal mesothelioma. However, the incorporation of locoregional therapy for PM in patients with PDAC has been poorly studied given the aggressive nature of pancreatic cancer and overall poor prognosis. With recent advances in existing treatment options, there may be a subset of patients who may derive benefits from locoregional control with cytoreduction and/or intraperitoneal chemotherapy. Critically, additional work is needed to determine PM-favorable clinical and tumoral predictive biomarkers to identify patients who may benefit from a more aggressive approach. We describe the current state of management of patients with peritoneal metastasis from PDAC and review the available data exploring peritoneal-directed therapy with cytoreductive surgery and/or intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Grace Wu
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Oliver J. Standring
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Daniel A. King
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | - Sepideh Gholami
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | - Craig E. Devoe
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | | | - Travis E. Grotz
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA (T.E.G.)
| | - Matthew J. Weiss
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | - Richard L. Whelan
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Danielle K. DePeralta
- Northwell Health, New Hyde Park, NY 11040, USA; (G.W.); (O.J.S.); (D.A.K.); (S.G.); (C.E.D.); (M.J.W.); (R.L.W.)
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| |
Collapse
|
2
|
Safari D, Fakhrolmobasheri M, Soleymanjahi S. Efficacy and safety of intraperitoneal chemotherapy for pancreatic cancer. BMC Surg 2024; 24:285. [PMID: 39367354 PMCID: PMC11451220 DOI: 10.1186/s12893-024-02526-9] [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: 06/25/2024] [Accepted: 08/07/2024] [Indexed: 10/06/2024] Open
Abstract
Pancreatic cancer is a highly aggressive cancer with unfavorable prognosis despite the therapeutic interventions. Intraperitoneal chemotherapy has recently shown potential outcomes in the presence of peritoneal metastases. However, a consensus is still lacking on different methods for intraperitoneal chemotherapy in pancreatic cancer. A variety of drugs and doses via three types of intraperitoneal chemotherapy have been studied. The prognosis and treatment strategies for pancreatic ductal adenocarcinoma (PDAC) will be significantly influenced by peritoneal dissemination and resectability of the macroscopic disease. Normothermic intraperitoneal chemotherapy (NIPEC) has been used for the treatment of peritoneal metastases of pancreatic carcinomas. Intraperitoneal chemotherapy is often combined with systemic therapies or surgical procedures which may lead to favorable combination therapies such as cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a relatively new approach that provides a homogenous and deep penetration of the chemotherapy into the peritoneum by producing aerosols. The present study aims to review the literature for recent evidence on intraperitoneal chemotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Dorsa Safari
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Fakhrolmobasheri
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saeed Soleymanjahi
- Division of Gastroenterology, Mass General Brigham, Harvard School of Medicine, 101 S Huntington Ave, Boston, MA, 02130, USA.
| |
Collapse
|
3
|
Di Giorgio A, Ferracci F, Bagalà C, Carbone C, Salvatore L, Strippoli A, Attalla El Halabieh M, Abatini C, Alfieri S, Pacelli F, Tortora G. Combined Nabpaclitaxel pressurized intraPeritoneal aerosol chemotherapy with systemic Nabpaclitaxel-Gemcitabine chemotherapy for pancreatic cancer peritoneal metastases: protocol of single-arm, open-label, phase II trial (Nab-PIPAC trial). Pleura Peritoneum 2024; 9:121-129. [PMID: 39544430 PMCID: PMC11558173 DOI: 10.1515/pp-2024-0010] [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: 06/18/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024] Open
Abstract
Objectives Current therapies show limited efficacy against peritoneal metastases (PM) from pancreatic cancer. Pressurized intra-peritoneal aerosol chemotherapy (PIPAC) has emerged as a novel intraperitoneal drug delivery method. Recently, a dose-escalation study identified the safe dose of Nabpaclitaxel for PIPAC administration, an ideal intraperitoneal chemotherapy agent against pancreatic cancer. Combining systemic NabPaclitaxel-Gemcitabine with NabPaclitaxel-PIPAC may enhance disease control in pancreatic cancer patients with PM. Methods The Nab-PIPAC trial is a single-center, prospective, open-label, phase II study (ClinicalTrials.gov identifier: NCT05371223). Its primary goal is to evaluate the antitumor activity of the combined treatment based on Disease Control Rate (DCR) using RECISTv.1.1 criteria. Secondary objectives include feasibility, safety, pathological response, progression-free and overall survival, nutritional status, quality of life, pharmacokinetics of NabPaclitaxel-PIPAC, and PM molecular evolution via translational research. The treatment protocol consists of three courses, each with two cycles of intravenous NabPaclitaxel-Gemcitabine and one cycle of NabPaclitaxel-PIPAC, with standard metastatic pancreatic cancer doses for the former and 112.5 mg/m2 for the latter. Sample size follows Simon's two-stage design: 12 patients in stage one and 26 in stage two (80 % power, 0.1 alpha). Results Partial results will be available after first stage enrollment. Conclusions This trial aims to determine the antitumor efficacy and safety of combining NabPaclitaxel-PIPAC with systemic NabPaclitaxel-Gemcitabine in pancreatic cancer patients with PM.
Collapse
Affiliation(s)
- Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Ferracci
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cinzia Bagalà
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Science, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lisa Salvatore
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonia Strippoli
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Miriam Attalla El Halabieh
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Abatini
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sergio Alfieri
- Surgical Unit of Digestive Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Fabio Pacelli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
4
|
Koti S, Demyan L, Deutsch G, Weiss M. Surgery for Oligometastatic Pancreatic Cancer: Defining Biologic Resectability. Ann Surg Oncol 2024; 31:4031-4041. [PMID: 38502293 PMCID: PMC11076395 DOI: 10.1245/s10434-024-15129-8] [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: 12/14/2023] [Accepted: 02/17/2024] [Indexed: 03/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is most often metastatic at diagnosis. As systemic therapy continues to improve alongside advanced surgical techniques, the focus has shifted toward defining biologic, rather than technical, resectability. Several centers have reported metastasectomy for oligometastatic PDAC, yet the indications and potential benefits remain unclear. In this review, we attempt to define oligometastatic disease in PDAC and to explore the rationale for metastasectomy. We evaluate the existing evidence for metastasectomy in liver, peritoneum, and lung individually, assessing the safety and oncologic outcomes for each. Furthermore, we explore contemporary biomarkers of biological resectability in oligometastatic PDAC, including radiographic findings, biochemical markers (such as CA 19-9 and CEA), inflammatory markers (including neutrophil-to-lymphocyte ratio, C-reactive protein, and scoring indices), and liquid biopsy techniques. With careful consideration of existing data, we explore the concept of biologic resectability in guiding patient selection for metastasectomy in PDAC.
Collapse
Affiliation(s)
- Shruti Koti
- Department of General Surgery, Northwell Health, Queens, NY, USA.
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA.
| | - Lyudmyla Demyan
- Department of General Surgery, Northwell Health, Queens, NY, USA
| | - Gary Deutsch
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Matthew Weiss
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
5
|
Kryh-Jensen CG, Fristrup CW, Ainsworth AP, Detlefsen S, Mortensen MB, Pfeiffer P, Tarpgaard LS, Graversen M. What is long-term survival in patients with peritoneal metastasis from gastric, pancreatic, or colorectal cancer? A study of patients treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC). Pleura Peritoneum 2023; 8:147-155. [PMID: 38144215 PMCID: PMC10739291 DOI: 10.1515/pp-2023-0038] [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: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Objectives A definition of long-term survival (LTS) in patients with peritoneal metastasis (PM) from gastric cancer (GC), pancreatic cancer (PC) or colorectal cancer (CRC) treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC) is lacking. We aimed to define LTS and investigate characteristics and treatment response in patients who reached LTS in data from two prospective trials. Methods Retrospective study of patients with GC-, PC-, or CRC-PM from the prospective PIPAC-OPC1 and PIPAC-OPC2 studies. The definition of LTS was based on published systematic reviews and randomized controlled trials. LTS was defined at the time point where 25 % of the patients were alive in these studies. Histology based response was evaluated by the mean Peritoneal Regression Grading Score (PRGS) using biopsies obtained prior to PIPAC 3, and defined by a mean PRGS of ≤2.0 or a decrease of mean PRGS of ≥1, compared to baseline. Results LTS was defined at 21 (GC), 15 (PC), and 24 (CRC) months. Fifty-one (47.2 %) patients (nine GC, 17 PC, 25 CRC) reached LTS calculated from the date of PM diagnosis. All but one received palliative chemotherapy before PIPAC, and 37 % received bidirectional treatment. More than 90 % of the LTS patients had response according to PRGS. The mOS from PIPAC 1 was 23.3, 12.4, and 28.5 months for GC, PC, and CRC LTS patients. Conclusions Patients with PM from GC, PC, and CRC treated with systemic chemotherapy and PIPAC can reach LTS and most show histological response. Causality must be further investigated.
Collapse
Affiliation(s)
- Charlotte G. Kryh-Jensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Claus W. Fristrup
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Alan P. Ainsworth
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Michael B. Mortensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Per Pfeiffer
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Line S. Tarpgaard
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Martin Graversen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- OPEN – Odense Patient Data Explorative Network, Odense University Hospital, Region of Southern Denmark, Odense, Denmark
| |
Collapse
|
6
|
Ezanno AC, Malgras B, Conan PL, Aime A, Fawaz J, Picchi H, Doat S, Pocard M. Reasons for stopping Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC): A retrospective study to improve future patient selection. PLoS One 2023; 18:e0287785. [PMID: 38033087 PMCID: PMC10688840 DOI: 10.1371/journal.pone.0287785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/13/2023] [Indexed: 12/02/2023] Open
Abstract
To improve the prognosis and maintain quality of life in patients with peritoneal metastasis (PM), a novel treatment has been introduced-pressurized intraperitoneal aerosol chemotherapy (PIPAC). The majority of teams propose at least 3 PIPAC procedures. However, for many patients PIPAC is stopped after only one or two procedures. The aim of this study was to identify the reasons for stopping PIPAC after only one or two procedures and to establish a profile of poor candidates. This retrospective, multicenter cohort study included all patients who underwent PIPAC in three French expert centers between 2015 and 2021. A total of 268 PIPAC procedures were performed in 89 patients. Of them, 48.3% of patients underwent fewer than three procedures: 28.1% had one, 20.2% two and 51.7% three or more PIPAC procedures. The main reason for stopping PIPAC, regardless of the number of procedures, was disease progression, in 55.8% of cases. Other reasons for stopping PIPAC were non-access to the abdominal cavity (7.9%), conversion to cytoreductive surgery (13.5%), post-PIPAC adverse events (7.9%), patients' wishes (10.1%) and death (2.2%). In univariate analysis, patients who received fewer than three PIPACs less frequently had chemotherapy beforehand (91% vs 100%, p = 0.05), less frequently had bimodal treatment (70% vs 87%, p = 0.04), had more ascites (median 80 ml vs 50 ml, p = 0.05) and more frequently had carcinomatosic ascites (48.8% vs 23.9%, p < 0.01). Performing PIPAC alone in chemotherapy-naïve patients with ascites should be avoided.
Collapse
Affiliation(s)
- Anne-Cécile Ezanno
- Department of Digestive Surgery, Begin Military Teaching Hospital, Saint Mandé, France
| | - Brice Malgras
- Department of Digestive Surgery, Begin Military Teaching Hospital, Saint Mandé, France
- French Military Health Service Academy, Ecole du Val de Grâce, Paris, France
| | - Pierre-Louis Conan
- Department of Infectiology, Begin Military Teaching Hospital, Saint Mandé, France
| | - Adeline Aime
- Department of Digestive Surgery, Begin Military Teaching Hospital, Saint Mandé, France
| | - Jade Fawaz
- Department of Digestive Surgery, Begin Military Teaching Hospital, Saint Mandé, France
| | - Hugo Picchi
- Department of Oncology, Begin Military Teaching Hospital, Saint Mandé, France
| | - Solène Doat
- Department of Hepato Gastro Enterology, La Pitié Salpétrière Hospital, Paris, France
| | - Marc Pocard
- Department of Digestive Surgery, La Pitié Salpetrière Hospital, Paris, France
- INSERM, U965 CART Unit, Paris, France
| |
Collapse
|
7
|
Ezanno AC, Malgras B, Pocard M. Pressurized intraperitoneal aerosol chemotherapy, reasons for interrupting treatment: a systematic review of the literature. Pleura Peritoneum 2023; 8:45-53. [PMID: 37304159 PMCID: PMC10249753 DOI: 10.1515/pp-2023-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/23/2023] [Indexed: 06/13/2023] Open
Abstract
Objectives Pressurized intraperitoneal aerosol chemotherapy (PIPAC) gives encouraging results in the treatment of peritoneal metastasis (PM). The current recommendations require at least 3 sessions of PIPAC. However, some patients do not complete the full treatment course and stop after only 1 or 2 procedures, hence the limited benefit. A literature review was performed, with search terms including "PIPAC" and "pressurised intraperitoneal aerosol chemotherapy." Content Only articles describing the causes for premature termination of the PIPAC treatment were analysed. The systematic search identified 26 published clinical articles related to PIPAC and reporting causes for stopping PIPAC. Summary The series range from 11 to 144 patients, with a total of 1352 patients treated with PIPAC for various tumours. A total of 3088 PIPAC treatments were performed. The median number of PIPAC treatments per patient was 2.1, the median PCI score at the time of the first PIPAC was 19 and the number of patients who did not complete the recommended 3 sessions of PIPAC was 714 (52.8%). Disease progression was the main reason for early termination of the PIPAC treatment (49.1%). The other causes were death, patients' wishes, adverse events, conversion to curative cytoreductive surgery and other medical reasons (embolism, pulmonary infection, etc…). Outlook Further investigations are necessary to better understand the causes for interrupting PIPAC treatment and also improving the selection of patients who are most likely to benefit from PIPAC.
Collapse
Affiliation(s)
- Anne-Cecile Ezanno
- Department of digestive surgery, Begin Military Teaching Hospital, Saint Mandé, France
| | - Brice Malgras
- Department of digestive surgery, Begin Military Teaching Hospital, Saint Mandé, France
- French Military health Service Academy, Ecole du Val de Grâce, Paris, France
| | - Marc Pocard
- Department of digestive surgery, La Pitié Salpétrière Hospital, Paris, France
- INSERM, U965 Cart unit, Paris, France
| |
Collapse
|
8
|
Baggaley AE, Lafaurie GBRC, Tate SJ, Boshier PR, Case A, Prosser S, Torkington J, Jones SEF, Gwynne SH, Peters CJ. Pressurized intraperitoneal aerosol chemotherapy (PIPAC): updated systematic review using the IDEAL framework. Br J Surg 2022; 110:10-18. [PMID: 36056893 PMCID: PMC10364525 DOI: 10.1093/bjs/znac284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/28/2022] [Accepted: 07/19/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Alice E Baggaley
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, UK
| | | | - Sophia J Tate
- Department of Anaesthesia, Swansea Bay University Health Board, Swansea, UK
| | - Piers R Boshier
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, UK
| | - Amy Case
- Department of Cancer Services, Swansea Bay University Health Board, Swansea, UK
| | - Susan Prosser
- Department of Library Services, Swansea Bay University Health Board, Swansea, UK
| | - Jared Torkington
- Department of Surgery, University Hospital of Wales, Cardiff, UK
| | - Sadie E F Jones
- Department of Obstetrics and Gynaecology, University Hospital of Wales, Cardiff, UK
| | - Sarah H Gwynne
- Department of Cancer Services, Swansea Bay University Health Board, Swansea, UK
| | - Christopher J Peters
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, UK
| |
Collapse
|
9
|
Frassini S, Calabretto F, Granieri S, Fugazzola P, Viganò J, Fazzini N, Ansaloni L, Cobianchi L. Intraperitoneal chemotherapy in the management of pancreatic adenocarcinoma: A systematic review and meta-analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 48:1911-1921. [PMID: 35688711 DOI: 10.1016/j.ejso.2022.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/09/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Pancreatic cancer represents one of the leading causes of cancer-related death worldwide. Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC), normothermic intraperitoneal chemotherapy (NIPEC), and pressurized intraperitoneal aerosol chemotherapy (PIPAC) has been proven with curative intent mainly for other tumors and there is a lack of consensus regarding possible benefits also in pancreatic cancer. The present systematic review and meta-analysis aim to provide an up-to-date overview of the effectiveness and safety of intraperitoneal treatments in the management of pancreatic cancer. METHODS A systematic review of articles was conducted according to PRISMA and AMSTAR-2 guidelines. 11 studies were included in the analysis. RESULTS We included in our analysis 212 patients subdivided in three groups: 64 in the HIPEC group (57 with prophylactic intent and 7 with curative intent), 55 in the PIPAC group and 93 in the NIPEC group. Primary outcomes were represented by survival rates; we evidenced at an observation time of three years a survival of 24% in the HIPEC group (25.5% in the prophylactic arm and 6.2% in the curative arm), 5.3% in the PIPAC group and 7.9% in the NIPEC group. CONCLUSIONS HIPEC could be considered as a promising technique for prophylaxis and treatment of peritoneal metastasis (PM) in case of borderline resectable and locally advanced disease. Increased survival rates emerged without additional morbidity when surgical resection and CRS are possible. In addition, our data about PIPAC and NIPEC as palliative treatment in unresectable disease seems to identify more favorable survival rates compared to literature.
Collapse
Affiliation(s)
- Simone Frassini
- University of Pavia, Corso Str. Nuova, 65, 27100, Pavia, Italy; Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Francesca Calabretto
- University of Pavia, Corso Str. Nuova, 65, 27100, Pavia, Italy; Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Stefano Granieri
- General Surgery Unit, ASST Vimercate, Via Santi Cosma e Damiano, 10, 20871, Vimercate, Italy.
| | - Paola Fugazzola
- Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Jacopo Viganò
- Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Nicola Fazzini
- University of Pavia, Corso Str. Nuova, 65, 27100, Pavia, Italy; Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Luca Ansaloni
- University of Pavia, Corso Str. Nuova, 65, 27100, Pavia, Italy; Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Lorenzo Cobianchi
- University of Pavia, Corso Str. Nuova, 65, 27100, Pavia, Italy; Unit of General Surgery I, Fondazione I.R.C.C.S. Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| |
Collapse
|
10
|
Satoi S, Takahara N, Fujii T, Isayama H, Yamada S, Tsuji Y, Miyato H, Yamaguchi H, Yamamoto T, Hashimoto D, Yamaki S, Nakai Y, Saito K, Baba H, Watanabe T, Ishii S, Hayashi M, Kurimoto K, Shimada H, Kitayama J. Synopsis of a clinical practice guideline for pancreatic ductal adenocarcinoma with peritoneal dissemination in Japan; Japan Peritoneal Malignancy Study Group. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2022; 29:600-608. [PMID: 34855287 PMCID: PMC9306579 DOI: 10.1002/jhbp.1085] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022]
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) with peritoneal dissemination have a dismal prognosis because discontinuation of systemic chemotherapy is required for massive ascites or poor performance status. The natural history, diagnosis and treatment of PDAC with peritoneal dissemination have not been fully investigated. We systematically reviewed published information on the clinical diagnosis and treatment of PDAC with peritoneal dissemination using the PubMed database (2000-2020) and provided recommendations in response to clinical questions. This guideline was created according to the "Minds Clinical Practice Guideline Development Guide 2017". The literature quality and body of evidence were evaluated with the GRADE System and classified into four levels ("strong", "medium", "weak", "very weak"). The strength of each final recommendation was decided by a vote of committee members based on the GRADE Grid method. These guidelines address three subjects: diagnostic, chemotherapeutic, and surgical approaches. They include nine clinical questions and statements with recommendation strengths, evidence levels, and agreement rates, in addition to one "column". This is the English synopsis of the 2021 Japanese clinical practice guideline for PDAC with peritoneal dissemination. It summarizes the clinical evidence for the diagnosis and treatment of PDAC with peritoneal dissemination and provides future perspectives.
Collapse
Affiliation(s)
- Sohei Satoi
- Department of SurgeryKansai Medical UniversityHirakataJapan
- Division of Surgical OncologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Naminatsu Takahara
- Department of GastroenterologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Tsutomu Fujii
- Department of Surgery and ScienceFaculty of MedicineAcademic AssemblyUniversity of ToyamaToyamaJapan
| | - Hiroyuki Isayama
- Department of GastroenterologyGraduate School of MedicineJuntendo UniversityTokyoJapan
| | - Suguru Yamada
- Department of SurgeryNagoya Central HospitalNagoyaJapan
| | - Yasushi Tsuji
- Department of Medical OncologyTonan HospitalSapporoJapan
| | - Hideyo Miyato
- Department of Gastrointestinal SurgeryJichi Medical UniversityShimotsukeJapan
| | | | | | | | - So Yamaki
- Department of SurgeryKansai Medical UniversityHirakataJapan
| | - Yousuke Nakai
- Department of GastroenterologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Kei Saito
- Department of GastroenterologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Hayato Baba
- Department of Surgery and ScienceFaculty of MedicineAcademic AssemblyUniversity of ToyamaToyamaJapan
| | - Toru Watanabe
- Department of Surgery and ScienceFaculty of MedicineAcademic AssemblyUniversity of ToyamaToyamaJapan
| | - Shigeto Ishii
- Department of GastroenterologyGraduate School of MedicineJuntendo UniversityTokyoJapan
| | - Masamichi Hayashi
- Department of Surgery IINagoya University Graduate School of MedicineNagoyaJapan
| | - Keisuke Kurimoto
- Department of Surgery IINagoya University Graduate School of MedicineNagoyaJapan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery and Clinical OncologyToho University Graduate School of MedicineTokyoJapan
| | - Joji Kitayama
- Department of Gastrointestinal SurgeryJichi Medical UniversityShimotsukeJapan
| |
Collapse
|
11
|
Rotolo S, Di Giorgio A, Cintoni M, Rinninella E, Palombaro M, Pulcini G, Schena CA, Chiantera V, Vizzielli G, Gasbarrini A, Pacelli F, Mele MC. Body composition and immunonutritional status in patients treated with pressurized intraperitoneal aerosol chemotherapy (PIPAC) for gastrointestinal peritoneal metastases: a prospective single-center analysis. Pleura Peritoneum 2022; 7:9-17. [PMID: 35602923 PMCID: PMC9069500 DOI: 10.1515/pp-2021-0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/02/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objectives
Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a novel drug administration method with promising efficacy for the treatment of peritoneal metastases (PM). This study aimed to evaluate the prognostic value of an immunonutritional assessment on the feasibility, safety, and survival in this setting.
Methods
Data of PM patients undergoing PIPAC between September 2018 and May 2020 were prospectively recorded. A CT scan-derived body composition assessment was performed for each patient.
Results
Fifty-one patients were enrolled, of which 30 (58%) underwent multiple PIPAC cycles, with a pathological response rate of 55%. Prognostic nutritional index (PNI) and neutrophil-to-lymphocytes predicted completion of more than one PIPAC cycle, with a cut off of 36.5 and 4.8 respectively. Muscle attenuation and body fat tissues were associated with pathological response. At multivariate Cox regression analysis, only the presence of a low PNI (HR 2.41, 95% CI 1.08–5.46) was significantly associated with a worse OS.
Conclusions
A pretreatment immunonutritional assessment may provide valuable information for PIPAC patients’ selection and survival, while body composition parameters are able to predict pathological response. Further larger studies are needed to validate the role of these biomarkers in tailoring the treatment and monitoring PM patients undergoing PIPAC.
Collapse
Affiliation(s)
- Stefano Rotolo
- Dipartimento di Scienze Mediche e Chirurgiche , UOC Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche (Di.Chir.On.S.) , Università degli Studi di Palermo , Palermo , Italy
| | - Andrea Di Giorgio
- Dipartimento di Scienze Mediche e Chirurgiche , UOC Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Marco Cintoni
- Dipartimento di Scienze Mediche e Chirurgiche , UOC di Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Emanuele Rinninella
- Dipartimento di Scienze Mediche e Chirurgiche , UOC di Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Marta Palombaro
- Dipartimento di Scienze Mediche e Chirurgiche , UOSD di Nutrizione Avanzata in Oncologia, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Gabriele Pulcini
- Dipartimento di Scienze Mediche e Chirurgiche , UOSD di Nutrizione Avanzata in Oncologia, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Carlo Alberto Schena
- Dipartimento di Scienze Mediche e Chirurgiche , UOC Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Vito Chiantera
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche (Di.Chir.On.S.) , Università degli Studi di Palermo , Palermo , Italy
| | - Giuseppe Vizzielli
- Dipartimento per la salute della Donna e del Bambino e della Salute Pubblica , UOC Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Antonio Gasbarrini
- Dipartimento di Scienze Mediche e Chirurgiche , UOC Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
- Dipartimento di Medicina e Chirurgia Traslazionale , Università Cattolica Del Sacro Cuore , Rome , Italy
| | - Fabio Pacelli
- Dipartimento di Scienze Mediche e Chirurgiche , UOC Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Maria Cristina Mele
- Dipartimento di Scienze Mediche e Chirurgiche , UOSD di Nutrizione Avanzata in Oncologia, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
- Dipartimento di Medicina e Chirurgia Traslazionale , Università Cattolica Del Sacro Cuore , Rome , Italy
| |
Collapse
|
12
|
Inzani F, Arciuolo D, Angelico G, Santoro A, Travaglino A, D'Alessandris N, Scaglione G, Valente M, Cianfrini F, Raffone A, Zannoni GF. Assessing Post-Treatment Pathologic Tumor Response in Female Genital Tract Carcinomas: An Update. Front Oncol 2022; 12:814989. [PMID: 35223496 PMCID: PMC8866564 DOI: 10.3389/fonc.2022.814989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/05/2022] [Indexed: 01/05/2023] Open
Abstract
In the last decades, several new therapeutic strategies have been introduced in the field of gynecologic oncology. These include neoadjuvant chemotherapy for high-grade serous tubo-ovarian carcinoma, hormonal fertility-sparing strategies for endometrial cancer, pressurized intraperitoneal aerosol chemotherapy (PIPAC) for surgically incurable peritoneal metastasis, and neoadjuvant treatments for locally advanced cervical carcinomas. All these recent advances lead to the development of novel scoring systems for the evaluation of pathological response related to specific treatments. In this regard, pathological evaluation of the morphological modifications related to these treatments and the definition of a tumor regression grading score have been introduced in clinical practice in order to achieve a more efficient prognostic stratification of patients affected by gynecological malignancies. The aim of the present paper is to provide a detailed review on the post-treatment pathological scoring systems in patients affected by gynecological malignancies.
Collapse
Affiliation(s)
- Frediano Inzani
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Damiano Arciuolo
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giuseppe Angelico
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angela Santoro
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Travaglino
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nicoletta D'Alessandris
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulia Scaglione
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michele Valente
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Cianfrini
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Raffone
- Gynecology and Obstetrics Unit, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy.,Division of Gynaecology and Human Reproduction Physiopathology, Department of Medical and Surgical Sciences (DIMEC), IRCCS Azienda Ospedaliero-Univeristaria di Bologna. S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Gian Franco Zannoni
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
13
|
Evaluation of the environmental contamination and exposure risk in medical/non-medical staff after oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy. Toxicol Appl Pharmacol 2021; 429:115694. [PMID: 34428445 DOI: 10.1016/j.taap.2021.115694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022]
Abstract
Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a technique to directly deliver chemotherapeutic drugs in the abdomen for the treatment of peritoneal metastases. Pressurization improves the treatment efficacy but increases the risk of exposure for the medical/non-medical staff who can be exposed by dermal or ocular contact, or inhalation of aerosols containing the cytotoxic drugs. The aim of this study was to evaluate the risk of exposure for the medical/non-medical staff (nurses, surgeons, anaesthesiologists and cleaning personnel; n = 13) during PIPAC with oxaliplatin performed according to the protocol recommended in France. Blood samples were collected 1 h before and immediately after PIPAC, and urine samples 1 h before, and then 3 h and the morning after PIPAC. In the control, non-exposed group (n = 7), only one urine and blood sample were collected. Surface contamination in the operating room was assessed in water- and Surfanios-impregnated wipe samples. The total elemental platinum in each sample was quantified by inductively coupled plasma mass spectrometry, using a method adapted to quantify trace amounts (ng.L-1) in very low volumes (100 μl). No surface contamination was detected. Although 25% of urine samples in the exposed group contained platinum, no statistical difference was observed in urine and plasma samples collected before and after PIPAC and with the control group samples. These findings suggest that the French PIPAC protocol does not increase the risk of exposure to platinum in all staff categories involved. This protocol could be considered in future occupational policies and consensus statements. Trial registration: NCT04014426.
Collapse
|
14
|
Rodolfino E, Di Marco M, Ilot A, Iezzi R, Gui B, Avesani G, Panico C, Strippoli A, Di Giorgio A, Pacelli F, Manfredi R. Radiologist Checklist for Selecting Patients to Undergo PIPAC (Pressurized IntraPeritoneal Aerosol Chemotherapy). Life (Basel) 2021; 11:life11090941. [PMID: 34575093 PMCID: PMC8472130 DOI: 10.3390/life11090941] [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: 06/20/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/03/2022] Open
Abstract
Peritoneal carcinomatosis frequently occurs in advanced gastrointestinal and gynecological cancers. As factors such as poor drug uptake and distribution cause chemotherapy to be less effective, alternative therapies have been explored. Introduced in 2013, PIPAC (pressurized intraperitoneal aerosol chemotherapy) uses aerosolized chemotherapeutics sprayed into the patient’s peritoneal cavity using a laparoscopic approach. Despite the literature showing encouraging data regarding the tolerability and efficacy of PIPAC, there is a lack of articles on the role that imaging plays in selecting patients suitable for PIPAC. The aim of this study is to combine literature-based evidence and clinical experience to provide information able to support training radiologists, as well as experienced radiologists interested in innovative therapies.
Collapse
Affiliation(s)
- Elena Rodolfino
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (E.R.); (B.G.); (G.A.); (C.P.); (R.M.)
| | - Margo’ Di Marco
- Section of Radiology, Department of Radiological and Hematological Scienses, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.M.); (A.I.)
| | - Alessia Ilot
- Section of Radiology, Department of Radiological and Hematological Scienses, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.M.); (A.I.)
| | - Roberto Iezzi
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (E.R.); (B.G.); (G.A.); (C.P.); (R.M.)
- Section of Radiology, Department of Radiological and Hematological Scienses, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.M.); (A.I.)
- Correspondence:
| | - Benedetta Gui
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (E.R.); (B.G.); (G.A.); (C.P.); (R.M.)
| | - Giacomo Avesani
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (E.R.); (B.G.); (G.A.); (C.P.); (R.M.)
| | - Camilla Panico
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (E.R.); (B.G.); (G.A.); (C.P.); (R.M.)
| | - Antonia Strippoli
- Comprehensive Cancer Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.D.G.); (F.P.)
| | - Fabio Pacelli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.D.G.); (F.P.)
| | - Riccardo Manfredi
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (E.R.); (B.G.); (G.A.); (C.P.); (R.M.)
- Section of Radiology, Department of Radiological and Hematological Scienses, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.M.); (A.I.)
| |
Collapse
|
15
|
Maier CF, Zhu L, Nanduri LK, Kühn D, Kochall S, Thepkaysone ML, William D, Grützmann K, Klink B, Betge J, Weitz J, Rahbari NN, Reißfelder C, Schölch S. Patient-Derived Organoids of Cholangiocarcinoma. Int J Mol Sci 2021; 22:ijms22168675. [PMID: 34445380 PMCID: PMC8395494 DOI: 10.3390/ijms22168675] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 12/16/2022] Open
Abstract
Cholangiocarcinoma (CC) is an aggressive malignancy with an inferior prognosis due to limited systemic treatment options. As preclinical models such as CC cell lines are extremely rare, this manuscript reports a protocol of cholangiocarcinoma patient-derived organoid culture as well as a protocol for the transition of 3D organoid lines to 2D cell lines. Tissue samples of non-cancer bile duct and cholangiocarcinoma were obtained during surgical resection. Organoid lines were generated following a standardized protocol. 2D cell lines were generated from established organoid lines following a novel protocol. Subcutaneous and orthotopic patient-derived xenografts were generated from CC organoid lines, histologically examined, and treated using standard CC protocols. Therapeutic responses of organoids and 2D cell lines were examined using standard CC agents. Next-generation exome and RNA sequencing was performed on primary tumors and CC organoid lines. Patient-derived organoids closely recapitulated the original features of the primary tumors on multiple levels. Treatment experiments demonstrated that patient-derived organoids of cholangiocarcinoma and organoid-derived xenografts can be used for the evaluation of novel treatments and may therefore be used in personalized oncology approaches. In summary, this study establishes cholangiocarcinoma organoids and organoid-derived cell lines, thus expanding translational research resources of cholangiocarcinoma.
Collapse
Affiliation(s)
- Christopher Fabian Maier
- Junior Clinical Cooperation Unit Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (C.F.M.); (L.Z.)
- Department of Surgery, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, 68167 Mannheim, Germany; (N.N.R.); (C.R.)
| | - Lei Zhu
- Junior Clinical Cooperation Unit Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (C.F.M.); (L.Z.)
- Department of Surgery, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, 68167 Mannheim, Germany; (N.N.R.); (C.R.)
| | - Lahiri Kanth Nanduri
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.K.N.); (D.K.); (S.K.); (M.-L.T.); (J.W.)
| | - Daniel Kühn
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.K.N.); (D.K.); (S.K.); (M.-L.T.); (J.W.)
| | - Susan Kochall
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.K.N.); (D.K.); (S.K.); (M.-L.T.); (J.W.)
| | - May-Linn Thepkaysone
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.K.N.); (D.K.); (S.K.); (M.-L.T.); (J.W.)
| | - Doreen William
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT) Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (K.G.); (B.K.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Konrad Grützmann
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT) Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (K.G.); (B.K.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Barbara Klink
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT) Partner Site Dresden, 01307 Dresden, Germany; (D.W.); (K.G.); (B.K.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center of Genetics, Laboratoire National de Santé (LNS), 3555 Dudelange, Luxembourg
| | - Johannes Betge
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models (B440), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Department of Medicine II, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Jürgen Weitz
- Department of Gastrointestinal, Thoracic and Vascular Surgery, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.K.N.); (D.K.); (S.K.); (M.-L.T.); (J.W.)
| | - Nuh N. Rahbari
- Department of Surgery, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, 68167 Mannheim, Germany; (N.N.R.); (C.R.)
| | - Christoph Reißfelder
- Department of Surgery, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, 68167 Mannheim, Germany; (N.N.R.); (C.R.)
| | - Sebastian Schölch
- Junior Clinical Cooperation Unit Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (C.F.M.); (L.Z.)
- Department of Surgery, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, 68167 Mannheim, Germany; (N.N.R.); (C.R.)
- Correspondence:
| |
Collapse
|
16
|
Lurvink RJ, Van der Speeten K, Rovers KP, de Hingh IHJT. The emergence of pressurized intraperitoneal aerosol chemotherapy as a palliative treatment option for patients with diffuse peritoneal metastases: a narrative review. J Gastrointest Oncol 2021; 12:S259-S270. [PMID: 33968442 DOI: 10.21037/jgo-20-497] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is an emerging palliative treatment for patients with unresectable peritoneal metastases. Potential advantages of PIPAC over current treatment options are a homogeneous intraperitoneal distribution, low local and systemic toxicity, and enhanced tumour penetration. Given these possible benefits, PIPAC is increasingly implemented in many centres worldwide. Scientific research into PIPAC is currently available from in vitro/in vivo/in animal studies, retrospective cohorts in humans, and phase I and II studies in humans. There are no results from randomised trials comparing PIPAC with conventional treatment, such as palliative systemic therapy. This narrative review aimed to provide an overview of the currently available literature on PIPAC. In general, repetitive PIPAC was feasible and safe for patients and operating room personnel. Primary and secondary non-access rates varied from 0-17% and 0-15%, respectively. Iatrogenic bowel injury was observed in 0-3% of PIPAC procedures. CTCAE grade 1-2 complications were common, mostly consisting of abdominal pain, nausea, vomiting, and fatigue. CTCAE grade 3-4 complications were uncommon, occurring on 0-15% of PIPAC procedures. Post-operative mortality rates of 0-2% were reported. The risk of occupational exposure to cytotoxic drugs was very low when strict safety guidelines were followed. Clinical heterogeneity was high in most studies, since, in general, patients with unresectable peritoneal metastases from a variety of primary tumours were included. Also, patients received either PIPAC monotherapy or PIPAC combined with concomitant systemic therapy, and were able to receive PIPAC in any line of palliative treatment. Since the results were generally not stratified for these three important factors, this severely complicates the interpretation of results. Based on the current literature, PIPAC may be regarded as a promising palliative treatment option in patients with diffuse peritoneal metastases. Initial results show that it is feasible and safe. However, well designed and (ideally) randomized controlled trials are urgently needed to determine the additional value of PIPAC in this setting. Until then, PIPAC should preferably be performed in the setting of clinical trials.
Collapse
Affiliation(s)
- Robin J Lurvink
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | | | - Koen P Rovers
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands.,GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
17
|
Rotolo S, Ferracci F, Santullo F, Lodoli C, Inzani F, Abatini C, Pacelli F, Di Giorgio A. Systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC): A case report of a multimodal treatment for peritoneal metastases of pancreatic origin. Int J Surg Case Rep 2020; 77S:S75-S78. [PMID: 33191190 PMCID: PMC7876684 DOI: 10.1016/j.ijscr.2020.10.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Integration of PIPAC to systemic chemotherapy can have good tolerance profile. Repeated biopsies performed during PIPAC allowed therapy-response monitoring. Analysis of biopsies performed during PIPAC allowed tailoring of chemotherapy. Integration of PIPAC to systemic chemotherapy can permit good quality of life.
Introduction Pancreatic ductal adenocarcinoma (PDAC) with peritoneal metastases (PM) has a dismal prognosis and palliative systemic chemotherapy, which represents the standard treatment option, has significant pharmacokinetics limitations and low efficacy. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a new method of drug delivery that is expected to maximize exposure of peritoneal nodules to antiblastic agents. A combination of systemic chemotherapy and PIPAC may be valuable. Presentation of case A 55 years old male affected by PDAC with synchronous PM underwent a multimodal treatment comprising systemic chemotherapy and PIPAC without any procedural-related adverse events. Tumor genomic profiling evaluation from peritoneal biopsies addressed further tailored systemic chemotherapy. Discussion The presented case illustrates the possibility of adding PIPAC to systemic chemotherapy with a fair tolerance profile and good quality of life while allowing monitoring of therapy-response and tailoring of the antiblastic treatment.
Collapse
Affiliation(s)
- Stefano Rotolo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Federica Ferracci
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Francesco Santullo
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Lodoli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Frediano Inzani
- Institute of Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Abatini
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Pacelli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|