1
|
Rietveld PCS, Guchelaar NAD, Sassen SDT, Koch BCP, Mathijssen RHJ, Koolen SLW. A Clinical Pharmacological Perspective on Intraperitoneal Chemotherapy. Drugs 2025:10.1007/s40265-025-02195-9. [PMID: 40411722 DOI: 10.1007/s40265-025-02195-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/26/2025]
Abstract
Peritoneal metastases (PM), frequently observed in malignancies such as ovarian, colorectal, pancreatic, and gastric cancers, present a significant therapeutic challenge due to poor prognosis and limited effectiveness to systemic chemotherapy. The peritoneal-plasma barrier reduces effective drug transfer from plasma to the peritoneal cavity, reducing cytotoxic effects on PM. Intraperitoneal (IP) chemotherapy offers a locoregional approach, enabling high local drug concentrations that can enhance therapeutic efficacy while limiting systemic toxicity. The three major methods for IP administration-hyperthermic intraperitoneal chemotherapy (HIPEC), pressurized intraperitoneal aerosol chemotherapy (PIPAC), and catheter-based IP (CBIP) chemotherapy-each provide unique pharmacokinetic (PK) advantages for PM treatment. This review provides a comprehensive update on the pharmacological rationale of IP chemotherapy, focusing on drug characteristics that support extended IP retention and effective tumor targeting. The effects of administration variables are discussed, highlighting their role in optimizing IP drug exposure. Additionally, recent PK data on commonly used drugs in IP therapy, including platinum-based agents, taxanes, and novel nanoparticle formulations, will be evaluated. While PK rationale supports the administration of IP chemotherapy, further efficacy results from ongoing clinical trials are still awaited. Innovations in nanoparticle-based formulations and controlled-release systems offer substantial potential for improving both drug retention and targeted delivery, enhancing treatment precision and minimizing systemic toxicity. Continued exploration in these areas, along with optimization of IP administration protocols, is vital for advancing patient outcomes, refining therapeutic strategies, and maximizing the benefits of IP chemotherapy in clinical practice.
Collapse
Affiliation(s)
- Pascale C S Rietveld
- Department of Clinical Pharmacy, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Sebastiaan D T Sassen
- Department of Clinical Pharmacy, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Clinical Pharmacy, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Clinical Pharmacy, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Brown ZJ, Krell M, Gitlin S, Ranjbar S, Vega DA, Pawlik TM. Prospects of the surgical management of colorectal peritoneal metastasis. J Gastrointest Surg 2025; 29:101940. [PMID: 39746646 DOI: 10.1016/j.gassur.2024.101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignancy with the propensity to metastasize. Common sites of metastasis include the liver, lungs, and peritoneum, with peritoneal metastases (PMs) having the worst prognosis. Unfortunately, systemic chemotherapy is often less effective in the treatment of PMs. Therefore, removal of all visible tumor via cytoreductive surgery along with intraperitoneal (IP) therapies has been used. METHODS A comprehensive review of the literature was conducted using MEDLINE/PubMed and Web of Science with an end date of September 1, 2024, regarding cytoreductive surgery and heated IP chemotherapy for CRC PMs. RESULTS Recent studies have called into question the utility of IP chemotherapy in the treatment of CRC PMs. However, regardless of IP chemotherapy, cytoreductive surgery has demonstrated an additional survival benefit for patients with PM secondary to CRC. DISCUSSION This study reviews the pathophysiology of CRC PM, the current treatment paradigms, and a pathway for improving outcomes in patients with CRC PM.
Collapse
Affiliation(s)
- Zachary J Brown
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States.
| | - Matthew Krell
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Saige Gitlin
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Suedeh Ranjbar
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Diego Alvarez Vega
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Timothy M Pawlik
- Division of Surgical Oncology, Department of Surgery, Wexner Medical Center and James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
3
|
Yılmaz E, Kacaroglu D, Ozden AK, Aydogan N. Gold nanoparticles decorated FOLFIRINOX loaded liposomes for synergistic therapy of pancreatic cancer. Int J Pharm 2025; 669:125067. [PMID: 39672312 DOI: 10.1016/j.ijpharm.2024.125067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Pancreatic cancer is predicted to be the second highest cause of cancer deaths by 2030, with a mortality rate of 98 % and a 5-year survival rate of only 4-8 %. FOLFIRINOX which consists of four main ingredients has shown superior efficacy in treating patients with pancreatic cancer compared to other agents and combinations. However, toxicities have prevented full-dose use of FOLFIRINOX. In this study, we present the design of a liposome nanosystem that enables the sequential release of a drug combination that is called FOLFIRINOX using lipid-based nanosystem synergistic chemo/photothermal therapy approaches. The co-eccentric liposome allowed us to locate the drug molecules in different locations giving us the flexibility to release them in a selected order. Core liposome (L2) has a melting temperature of 53.63 °C, it was decorated by gold nanoparticle (L2@AuNP) to bring photothermal responsiveness. The outer liposome structure had a lower melting temperature, which facilitated the sequential release process. The efficacy of photothermal therapy for nanosystem was calculated. The results indicate that coating L2@AuNP nanostructure with L1 liposomes improves efficacy by stabilizing gold nanoparticles. FOLFIRINOX components are encapsulated in a concentric liposome structure according to the order of administration into the body. The concentric liposome structure enables the sequential release of multiple drugs due to the varying phase transition temperatures of the liposomes. The cytotoxic effect of these formulations was evaluated on Panc-1 pancreatic cancer cells; the lowest cell viability was obtained in 4 Liposome(L) under 5 min NIR irradiation. Combination therapy has a higher therapeutic efficacy (70.45 %) when compared to chemotherapy and photothermal therapy used separately. The study's results show the potential of combination therapies to improve therapeutic outcomes, providing a promising path for future research and clinical application.
Collapse
Affiliation(s)
- Emine Yılmaz
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey
| | - Demet Kacaroglu
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Ayse Kevser Ozden
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Nihal Aydogan
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey; Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara, 06800, Turkey.
| |
Collapse
|
4
|
Namakshenas P, Crezee J, Tuynman JB, Tanis PJ, Oei AL, Kok HP. Computational Evaluation of Improved HIPEC Drug Delivery Kinetics via Bevacizumab-Induced Vascular Normalization. Pharmaceutics 2025; 17:155. [PMID: 40006522 PMCID: PMC11859678 DOI: 10.3390/pharmaceutics17020155] [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: 12/23/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Oxaliplatin-based hyperthermic intraperitoneal chemotherapy (HIPEC) using the original 30 min protocol has shown limited benefits in patients with peritoneal metastasis of colorectal cancer (PMCRC), likely due to the short duration, which limits drug penetration into tumor nodules. Bevacizumab, an antiangiogenic antibody that modifies the tumor microenvironment, may improve drug delivery during HIPEC. This in silico study evaluates the availability of oxaliplatin within tumor nodules when HIPEC is performed after bevacizumab treatment. Methods: Using a computational fluid dynamics (CFD) model of HIPEC, the temperature and oxaliplatin distribution within the rat abdomen were calculated, followed by a model of drug transport within tumor nodules located at various sites in the peritoneum. The vascular normalization effect of the bevacizumab treatment was incorporated by adjusting the biophysical parameters of the tumor nodules. The effective penetration depth values, including the thermal enhancement ratio of cytotoxicity, were then compared between HIPEC alone and HIPEC combined with the bevacizumab treatment. Results: After bevacizumab treatments at doses of 0.5 mg/kg and 5 mg/kg, the oxaliplatin availability increased by up to 20% and 45% when HIPEC was performed during the vascular normalization phase, with the penetration depth increasing by 1.5-fold and 2.3-fold, respectively. Tumors with lower collagen densities and larger vascular pore sizes showed higher oxaliplatin enhancement after the combined treatment. Bevacizumab also enabled a reduction in the oxaliplatin dose (up to half at 5 mg/kg bevacizumab) while maintaining effective drug levels in the tumor nodules, potentially reducing systemic toxicity. Conclusions: These findings suggest that administering oxaliplatin-based HIPEC during bevacizumab-induced vascular normalization could significantly improve drug penetration and enhance treatment efficacy.
Collapse
Affiliation(s)
- Pouya Namakshenas
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
| | - Jurriaan B. Tuynman
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Pieter J. Tanis
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Arlene L. Oei
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
| | - H. Petra Kok
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
| |
Collapse
|
5
|
Morera-Ocon FJ, Navarro-Campoy C, Guastella T, Landete-Molina F. Controversies around the treatment of peritoneal metastases of colorectal cancer. World J Gastrointest Oncol 2025; 17:100199. [PMID: 39817141 PMCID: PMC11664603 DOI: 10.4251/wjgo.v17.i1.100199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
In this editorial we examine the article by Wu et al published in the World Journal of Gastrointestinal Oncology. Surgical resection for peritoneal metastases from colorectal cancer (CRC) has been gradually accepted in the medical oncology community. A randomized trial (PRODIGE 7) on cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) failed to prove any benefit of oxaliplatin in the overall survival of patients with peritoneal metastases from colorectal origin. Nevertheless, isolated systemic chemotherapy for CRC stage IV has demonstrated a reduced response in peritoneal metastases than that obtained in other metastatic sites such as the liver. Another tool is required in those patients to achieve more local control of the disease. Surgical groups in peritoneal surgery continue to use HIPEC in their procedures, using other agents than oxaliplatin for peritoneal cavity infusion, such as mitomycin C. These patients present with complex surgical issues to manage, and consequently a large burden of complications has to be anticipated. Therefore, identifying patients who will benefit from CRS with or without HIPEC would be of great interest.
Collapse
Affiliation(s)
| | - Clara Navarro-Campoy
- Department of Gynecology and Obstetrics, Hospital 9 Octubre, Valencia 46015, Spain
| | - Ticiano Guastella
- Department of Pathology, Hospital General de Requena, Requena 46340, Spain
| | | |
Collapse
|
6
|
Zubair M, Uddin I, Dickinson R, Diederich CJ. Delivering Volumetric Hyperthermia to Head and Neck Cancer Patient-Specific Models Using an Ultrasound Spherical Random Phased Array Transducer. Bioengineering (Basel) 2024; 12:14. [PMID: 39851288 PMCID: PMC11760898 DOI: 10.3390/bioengineering12010014] [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: 11/15/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
In exploring adjuvant therapies for head and neck cancer, hyperthermia (40-45 °C) has shown efficacy in enhancing chemotherapy and radiation, as well as the delivery of liposomal drugs. Current hyperthermia treatments, however, struggle to reach large deep tumors uniformly and non-invasively. This study investigates the feasibility of delivering targeted uniform hyperthermia deep into the tissue using a non-invasive ultrasound spherical random phased array transducer. Simulations in 3D patient-specific models for thyroid and oropharyngeal cancers assessed the transducer's proficiency. The transducer consisting of 256 elements randomly positioned on a spherical shell, operated at a frequency of 1 MHz with various phasing schemes and power modulations to analyze 40, 41, and 43 °C isothermal volumes and the penetration depth of the heating volume, along with temperature uniformity within the target area using T10, T50, and T90 temperatures, across different tumor models. Intensity distributions and volumetric temperature contours were calculated to define moderate hyperthermia boundaries. The results indicated the array's ability to produce controlled heating volumes from 1 to 48 cm3 at 40 °C, 0.35 to 27 cm3 at 41 °C, and 0.1 to 8 cm3 at 43 °C. The heating depths ranged from 7 to 39 mm minimum and 52 to 59 mm maximum, measured from the skin's inner surface. The transducer, with optimal phasing and water-cooled bolus, confined the heating to the targeted regions effectively. Multifocal sonications also improved the heating homogeneity, reducing the length-to-diameter ratio by 38% when using eight foci versus a single one. This approach shows potential for treating a range of tumors, notably deep-seated and challenging oropharyngeal cancers.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94115, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Imad Uddin
- Department of Cardiology, Hayat Abad Medical Complex, Peshawar 23301, Pakistan
| | - Robert Dickinson
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Chris J. Diederich
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94115, USA
| |
Collapse
|
7
|
Dong FH, Shan YQ, Kong WC, Wei HR, Zhou LP, Yang YB, Shi J, Ji CH, Zhang YJ. Hyperthermic intraperitoneal chemotherapy: Ideal and reality. Asian J Surg 2024:S1015-9584(24)02488-6. [PMID: 39567292 DOI: 10.1016/j.asjsur.2024.10.155] [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: 05/19/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Hyperthermic Intraperitoneal Chemoperfusion (HIPEC) is a new adjuvant therapy for the treatment of abdominal malignant tumors and metastases, which has shown great potential. The HIPEC technique can effectively kill residual lesions in the abdominal cavity through the synergistic sensitization effect of thermal chemotherapy and the circulating perfusion and washing effect of large volume perfusion fluid, thereby reducing the occurrence of malignant ascites and reducing the risk of postoperative recurrence and metastasis. However, there are still many problems in the practical operation of HIPEC, such as non-uniform distribution of perfusate temperature, inadequate perfusion due to the presence of 'dead space', incomplete cytoreductive surgery (CRS), instances of catheter obstruction during perfusate circulation, the lack of a uniform standard for selecting appropriate HIPEC techniques, occupational exposure of medical personnel during the HIPEC procedure, and the selection of HIPEC chemotherapy regimens for patients with various types of tumors. This article aims to summarize the problems encountered in the clinical practice of HIPEC, explore the problems that can be improved, and put forward some suggestions, so as to promote the further standardization and normalization of HIPEC technology.
Collapse
Affiliation(s)
- Fan-He Dong
- Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yu-Qiang Shan
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| | - Wen-Cheng Kong
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| | - Hao-Ran Wei
- Zhejiang Chinese Medical University, Hangzhou, China.
| | - Lin-Po Zhou
- Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ye-Bin Yang
- Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jing Shi
- Zhejiang Chinese Medical University, Hangzhou, China.
| | - Cheng-Hao Ji
- Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yi-Jia Zhang
- Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
8
|
Sleiman MJ, Jelip A, Buchs N, Toso C, Liot E, Koessler T, Meyer J, Meurette G, Ris F. Pressurized Intraperitoneal Aerosol Chemotherapy for Peritoneal Carcinomatosis in Colorectal Cancer Patients: A Systematic Review of the Evidence. Cancers (Basel) 2024; 16:3661. [PMID: 39518099 PMCID: PMC11544814 DOI: 10.3390/cancers16213661] [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: 09/30/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Pressurized intraperitoneal aerosol chemotherapy (PIPAC) consists of the administration of aerosolized chemotherapy into the abdominal cavity of patients suffering from peritoneal carcinomatosis. Our aim was to review the evidence supporting PIPAC in patients with peritoneal carcinomatosis from colorectal cancer. METHODS A systematic review was performed in accordance with the 2020 PRISMA guideline. MEDLINE and CENTRAL were searched using combinations of terms including "Peritoneal carcinomatosis", "Peritoneal metastasis", "PIPAC", "Pressurized intraperitoneal aerosol chemotherapy" and "Colorectal cancer". Original studies, in English, including patients treated with PIPAC for colorectal peritoneal carcinomatosis, were considered eligible. Case reports, non-English or French language articles and secondary analyses were excluded. RESULTS A total of 385 articles were screened and 374 articles were excluded, leaving 11 publications for inclusion in the qualitative analysis. The included studies totalized 949 patients who received PIPAC for peritoneal carcinomatosis due to colorectal cancer. The median peritoneal carcinomatosis index (PCI) ranged from 10 to 31. In all studies, the complete PIPAC protocol was achieved with an average of two to three 3 PIPAC sessions per patient. Oxaliplatin (OX) was used as a chemotherapeutic agent in all studies and could be associated with intravenous 5-FU and leucovorin. Most post-operative adverse events were recorded as mild to moderate with no intraoperative complications. Only four studies reported a decrease in the average PCI score for 50% of the patients. Median overall survival ranged from 8 to 37.8 months. Quality of life indicators were stable between PIPAC-OX cycles with a small but not statistically significant trend of improvement of most functional scales. CONCLUSIONS PIPAC for peritoneal carcinomatosis from colorectal origin is feasible, safe and tolerable. Its impact on survival outcomes or quality of life remains to be demonstrated by randomized trials.
Collapse
Affiliation(s)
- Marwan-Julien Sleiman
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| | - Annamaria Jelip
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| | - Nicolas Buchs
- Division of Digestive Surgery, Hôpital La Tour, 1217 Meyrin, Switzerland;
| | - Christian Toso
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| | - Emilie Liot
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| | - Thibaud Koessler
- Division of Oncology, University Hospitals of Geneva, 1205 Geneva, Switzerland;
| | - Jeremy Meyer
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| | - Guillaume Meurette
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| | - Frederic Ris
- Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland; (A.J.); (C.T.); (E.L.); (J.M.); (G.M.); (F.R.)
| |
Collapse
|
9
|
Rauwerdink P, van de Vlasakker VCJ, Wassenaar ECE, Rovers KP, Los M, Herbschleb KH, Creemers GJM, Thijs AMJ, Raicu MG, Huysentruyt CJR, van der Hoeven EJRJ, Nederend J, Peeters RYM, Deenen MJ, Elias SG, Fijneman RJA, Constantinides A, Kranenburg O, Burger PWA, Nienhuijs SW, Wiezer RJ, Lurvink RJ, de Hingh IHJT, Boerma D. First-line palliative systemic therapy alternated with oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy for unresectable colorectal peritoneal metastases: A single-arm phase II trial (CRC-PIPAC-II). EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108487. [PMID: 38905732 DOI: 10.1016/j.ejso.2024.108487] [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: 03/10/2024] [Revised: 05/22/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Palliative systemic therapy alternated with electrostatic precipitation oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy (ePIPAC) has never been prospectively investigated in patients with unresectable colorectal peritoneal metastases (CPM). The CRC-PIPAC-II study aimed to assess safety, feasibility and efficacy of such bidirectional therapy. METHODS This two-center, single-arm, phase II trial enrolled chemotherapy-naïve patients to undergo three treatment cycles, consisting of systemic therapy (CAPOX, FOLFOX, FOLFIRI, or FOLFOXIRI, all with bevacizumab) and oxaliplatin-based ePIPAC (92 mg/m2) with intravenous leucovorin (20 mg/m2) and 5-fluorouracil (400 mg/m2). Primary outcome were major treatment-related adverse events. Secondary outcomes included minor events, tumor response, progression-free survival (PFS) and overall survival (OS). RESULTS Twenty patients completed 52 treatment cycles. Fifteen major events occurred in 7 patients (35 %): 5 events (33 %) related to systemic therapy; 5 (33 %) related to ePIPAC; and 5 (33 %) were biochemical events. No treatment-related deaths occurred. All patients experienced minor events, mostly abdominal pain, nausea and peripheral sensory neuropathy. After treatment, radiological, pathological, cytological, and biochemical response was observed in 0 %, 88 %, 38 %, and 31 % of patients respectively. Curative surgery was achieved in one patient. Median PFS was 10.0 months (95 % confidence interval [CI] 8.0-13.0) and median OS was 17.5 months (95 % CI 13.0-not reached). CONCLUSIONS Combining palliative systemic therapy with oxaliplatin-based ePIPAC in patients with unresectable CPM was feasible and showed an acceptable safety profile. Treatment-induced response and survival are promising, yet further research is required to determine the additional value of ePIPAC to systemic therapy.
Collapse
Affiliation(s)
| | | | | | - Koen P Rovers
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Maartje Los
- Department of Medical Oncology, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Karin H Herbschleb
- Department of Medical Oncology, St. Antonius Hospital, Nieuwegein, Netherlands
| | | | | | - Mihaela G Raicu
- Department of Pathology DNA, St. Antonius Hospital, Nieuwegein, Netherlands
| | | | | | - Joost Nederend
- Department of Radiology, Catharina Hospital, Eindhoven, Netherlands
| | - Rifka Y M Peeters
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Sjoerd G Elias
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Remond J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alexander Constantinides
- Lab Translational Oncology, Division Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Onno Kranenburg
- Lab Translational Oncology, Division Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pim W A Burger
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | | | - René J Wiezer
- Department of Surgery, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Robin J Lurvink
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands; Department of Epidemiology, School for Oncology and Developmental Biology, GROW, Maastricht, Netherlands.
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, Netherlands.
| |
Collapse
|
10
|
Ahmed Eltayeb S, Dressler JM, Schlatt L, Pernecker M, Neugebauer U, Karst U, Ciarimboli G. Interaction of the chemotherapeutic agent oxaliplatin and the tyrosine kinase inhibitor dasatinib with the organic cation transporter 2. Arch Toxicol 2024; 98:2131-2142. [PMID: 38589558 PMCID: PMC11169033 DOI: 10.1007/s00204-024-03742-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
Oxaliplatin (OHP) is effective in colorectal cancer treatment but induces peripheral neurotoxicity (OHP-induced peripheral neurotoxicity, OIPN), diminishing survivor quality of life. Organic cation transporter 2 (OCT2) is a key OHP uptake pathway in dorsal root ganglia. Competing for OCT2-mediated OHP uptake, such as with the tyrosine kinase inhibitor dasatinib, may mitigate OHP side effects. We investigated OHP and dasatinib interaction with OCT2 in human embryonic kidney 293 (HEK293) cells expressing OCT2 within a 10-3 to 10-7 M concentration range. Uptake competition experiments using fluorescent organic cation 4-(4-dimethylaminostyryl)-N-methylpyridinium (ASP+, 1 µM) and mass spectrometry (MS) to determine cellular platinum content indicated that OHP (100 µM) is an OCT2 substrate, mediating OHP cellular toxicity. ASP+ and MS analysis revealed dasatinib as a non-transported inhibitor of hOCT2 (IC50 = 5.9 µM) and as a regulator of OCT2 activity. Dasatinib reduced transporter Vmax, potentially via Y544 phosphorylation suppression. MS analysis showed cellular dasatinib accumulation independent of hOCT2. Although 3 µM dasatinib reduced 100 µM OHP accumulation in hOCT2-HEK293 cells, co-incubation with dasatinib and OHP did not prevent OHP toxicity, possibly due to dasatinib-induced cell viability reduction. In summary, this study demonstrates OHP as an OCT2 substrate and dasatinib as a non-transported inhibitor and regulator of OCT2, offering potential for OIPN mitigation.
Collapse
Affiliation(s)
- Sara Ahmed Eltayeb
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1/A14, 48149, Münster, Germany
| | - Julia M Dressler
- Institut Für Anorganische Und Analytische Chemie, Universität Münster, Münster, Germany
| | - Lukas Schlatt
- Institut Für Anorganische Und Analytische Chemie, Universität Münster, Münster, Germany
| | - Moritz Pernecker
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1/A14, 48149, Münster, Germany
| | - Ute Neugebauer
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1/A14, 48149, Münster, Germany
| | - Uwe Karst
- Institut Für Anorganische Und Analytische Chemie, Universität Münster, Münster, Germany
| | - Giuliano Ciarimboli
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1/A14, 48149, Münster, Germany.
| |
Collapse
|
11
|
Rietveld PCS, Guchelaar NAD, van Eerden RAG, de Boer NL, de Bruijn P, Sassen SDT, Madsen EVE, Koch BCP, Verhoef C, Burger JWA, Mathijssen RHJ, Koolen SLW. Intraperitoneal pharmacokinetics of systemic oxaliplatin, 5-fluorouracil and bevacizumab in patients with colorectal peritoneal metastases. Biomed Pharmacother 2024; 176:116820. [PMID: 38810398 DOI: 10.1016/j.biopha.2024.116820] [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: 03/29/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Peritoneal metastases (PM) commonly occur in colorectal cancer patients. Systemic chemotherapy yields poor outcomes for these patients. It is hypothesised that traditional systemic chemotherapy is not very effective for this patient population. This study investigates to what extent systemic anti-cancer therapy crosses the peritoneal barrier. METHODS In a Phase I study, eighteen patients received systemic oxaliplatin, 5-FU, and bevacizumab. Plasma and peritoneal fluid samples were collected to measure drug concentrations. A non-compartmental analysis determined the Area Under the Curve (AUC) for oxaliplatin and 5-FU in both matrices. Intraperitoneal (IP) and intravenous (IV) exposure ratios were calculated, along with the bevacizumab concentration IP/IV ratio. The relationship between tumour load and IP/IV ratios and the correlation between the IP/IV ratios of different treatments were assessed statistically. RESULTS A total of 438 5-FU samples and 578 oxaliplatin samples were analysed in plasma and peritoneal fluid. Bevacizumab was quantified with 17 measurements in plasma and 15 measurements IP. Median IP/IV ratios were 0.143, 0.352 and 0.085 for 5-FU, oxaliplatin and bevacizumab, respectively. Oxaliplatin exhibited a longer IP half-life than 5-FU. A correlation was found between oxaliplatin and bevacizumab IP/IV ratios (R=0.69, p=0.01). No statistical correlations were found between the other investigated drugs. CONCLUSIONS Our findings indicate that only a small percentage of systemically administered anti-cancer treatment reaches the IP cavity, questioning their efficacy against PM. This strengthens the hypothesis for repeated intraperitoneal chemotherapy to reach adequate anti-cancer drug levels.
Collapse
Affiliation(s)
- Pascale C S Rietveld
- Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands; Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, the Netherlands.
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ruben A G van Eerden
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Nadine L de Boer
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Peter de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sebastiaan D T Sassen
- Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, the Netherlands
| | - Eva V E Madsen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Birgit C P Koch
- Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jacobus W A Burger
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Stijn L W Koolen
- Department of Clinical Pharmacy, Erasmus MC, Rotterdam, the Netherlands; Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Li X, Liu G, Wu W. Progress in Biological Research and Treatment of Pseudomyxoma Peritonei. Cancers (Basel) 2024; 16:1406. [PMID: 38611084 PMCID: PMC11010892 DOI: 10.3390/cancers16071406] [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: 03/01/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Pseudomyxoma peritonei (PMP) is a rare disease characterized by extensive peritoneal implantation and mass secretion of mucus after primary mucinous tumors of the appendix or other organ ruptures. Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is currently the preferred treatment, with excellent efficacy and safety, and is associated with breakthrough progress in long-term disease control and prolonged survival. However, the high recurrence rate of PMP is the key challenge in its treatment, which limits the clinical application of multiple rounds of CRS-HIPEC and does not benefit from conventional systemic chemotherapy. Therefore, the development of alternative therapies for patients with refractory or relapsing PMP is critical. The literature related to PMP research progress and treatment was searched in the Web of Science, PubMed, and Google Scholar databases, and a literature review was conducted. The overview of the biological research, treatment status, potential therapeutic strategies, current research limitations, and future directions associated with PMP are presented, focuses on CRS-HIPEC therapy and alternative or combination therapy strategies, and emphasizes the clinical transformation prospects of potential therapeutic strategies such as mucolytic agents and targeted therapy. It provides a theoretical reference for the treatment of PMP and the main directions for future research.
Collapse
Affiliation(s)
- Xi Li
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guodong Liu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
13
|
Quénet F, Carrère S, Sgarbura O. [Contribution of intraperitoneal chemotherapy in the treatment of colorectal peritoneal carcinoma. HIPEC, PIPAC, state of the art and future directions]. Bull Cancer 2024; 111:285-290. [PMID: 38331695 DOI: 10.1016/j.bulcan.2023.10.006] [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: 07/18/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 02/10/2024]
Abstract
After more than a decade of good results using the combination of cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) in the treatment of peritoneal carcinosis of colorectal origin, the PRODIGE7 study, which specifically evaluated the role of HIPEC, failed to show any superiority in terms of overall and disease-free survival for the CRS+HIPEC combination compared with CRS alone. This study constituted a radical change in the knowledge and therapeutic attitudes observed to date. After reviewing the literature and the consensus of national and international experts, a synthesis is provided, together with an outlook on the questions raised and the therapeutic trials and innovations of the near future. An analysis of recent advances due to the advent of a new technique, PIPAC, is also proposed, as well as a review of current therapeutic trials in this field.
Collapse
Affiliation(s)
- François Quénet
- Service de chirurgie oncologique, ICM Montpellier, 208, avenue des Apothicaires, 34000 Montpellier, France.
| | - Sébastien Carrère
- Service de chirurgie oncologique, ICM Montpellier, 208, avenue des Apothicaires, 34000 Montpellier, France
| | - Olivia Sgarbura
- Service de chirurgie oncologique, ICM Montpellier, 208, avenue des Apothicaires, 34000 Montpellier, France
| |
Collapse
|
14
|
Bhatt A, Glehen O, Zivanovic O, Brennan D, Nadeau C, Van Driel W, Bakrin N. The 2022 PSOGI International Consensus on HIPEC Regimens for Peritoneal Malignancies: Epithelial Ovarian Cancer. Ann Surg Oncol 2023; 30:8115-8137. [PMID: 37561343 DOI: 10.1245/s10434-023-13932-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/27/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND AND AIM We report the results of an international consensus on hyperthermic intraperitoneal chemotherapy (HIPEC) regimens for epithelial ovarian cancer (EOC) performed with the following goals: To define the indications for HIPEC To identify the most suitable HIPEC regimens for each indication in EOC To identify areas of future research on HIPEC To provide recommendations for some aspects of perioperative care for HIPEC METHODS: The Delphi technique was used with two rounds of voting. There were three categories of questions: evidence-based recommendations [using the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) system with the patient, intervention, comparator, and outcome (PICO) method], an opinion survey, and research recommendations. RESULTS Seventy-three (67.5%) of 108 invited experts responded in round I, and 68 (62.9%) in round II. Consensus was achieved for 34/38 (94.7%) questions. However, a strong positive consensus that would lead to inclusion in routine care was reached for only 6/38 (15.7%) questions. HIPEC in addition to interval cytoreductive surgery (CRS) received a strong positive recommendation that merits inclusion in routine care. Single-agent cisplatin was the only drug recommended for routine care, and OVHIPEC-1 was the most preferred regimen. The panel recommended performing HIPEC for a minimum of 60 min with a recommended minimum intraabdominal temperature of 41°C. Nephroprotection with sodium thiosulfate should be used for cisplatin HIPEC. CONCLUSIONS The results of this consensus should guide clinical decisions on indications of HIPEC and the choice and various parameters of HIPEC regimens and could fill current knowledge gaps. These outcomes should be the basis for designing future clinical trials on HIPEC in EOC.
Collapse
Affiliation(s)
- Aditi Bhatt
- Department of Surgical Oncology, KD Hospital, Ahmedabad, India.
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier, Lyon-sud, Lyon, France
| | - Oliver Zivanovic
- Department of Gynecological Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donal Brennan
- UCD Gynaecological Oncology Group, UCD School of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Cedric Nadeau
- Department of Gynecological Oncology, CHU de Poitiers, Poitiers, Cedex, France
| | - Willemien Van Driel
- Department of Gynecological Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Naoual Bakrin
- Department of Surgical Oncology, Centre Hospitalier, Lyon-sud, Lyon, France
| |
Collapse
|
15
|
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: 2.5] [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
|
16
|
Xia W, Geng Y, Hu W. Peritoneal Metastasis: A Dilemma and Challenge in the Treatment of Metastatic Colorectal Cancer. Cancers (Basel) 2023; 15:5641. [PMID: 38067347 PMCID: PMC10705712 DOI: 10.3390/cancers15235641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 10/25/2024] Open
Abstract
Peritoneal metastasis (PM) is a common mode of distant metastasis in colorectal cancer (CRC) and has a poorer prognosis compared to other metastatic sites. The formation of PM foci depends on the synergistic effect of multiple molecules and the modulation of various components of the tumor microenvironment. The current treatment of CRC-PM is based on systemic chemotherapy. However, recent developments in local therapeutic modalities, such as cytoreductive surgery (CRS) and intraperitoneal chemotherapy (IPC), have improved the survival of these patients. This article reviews the research progress on the mechanism, characteristics, diagnosis, and treatment strategies of CRC-PM, and discusses the current challenges, so as to deepen the understanding of CRC-PM among clinicians.
Collapse
Affiliation(s)
- Wei Xia
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou 213003, China;
| | - Yiting Geng
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou 213003, China;
| | - Wenwei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou 213003, China;
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| |
Collapse
|
17
|
Cheng F, Zhang R, Sun C, Ran Q, Zhang C, Shen C, Yao Z, Wang M, Song L, Peng C. Oxaliplatin-induced peripheral neurotoxicity in colorectal cancer patients: mechanisms, pharmacokinetics and strategies. Front Pharmacol 2023; 14:1231401. [PMID: 37593174 PMCID: PMC10427877 DOI: 10.3389/fphar.2023.1231401] [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: 05/30/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023] Open
Abstract
Oxaliplatin-based chemotherapy is a standard treatment approach for colorectal cancer (CRC). However, oxaliplatin-induced peripheral neurotoxicity (OIPN) is a severe dose-limiting clinical problem that might lead to treatment interruption. This neuropathy may be reversible after treatment discontinuation. Its complicated mechanisms are related to DNA damage, dysfunction of voltage-gated ion channels, neuroinflammation, transporters, oxidative stress, and mitochondrial dysfunction, etc. Several strategies have been proposed to diminish OIPN without compromising the efficacy of adjuvant therapy, namely, combination with chemoprotectants (such as glutathione, Ca/Mg, ibudilast, duloxetine, etc.), chronomodulated infusion, dose reduction, reintroduction of oxaliplatin and topical administration [hepatic arterial infusion chemotherapy (HAIC), pressurized intraperitoneal aerosol chemotherapy (PIPAC), and hyperthermic intraperitoneal chemotherapy (HIPEC)]. This article provides recent updates related to the potential mechanisms, therapeutic strategies in treatment of OIPN, and pharmacokinetics of several methods of oxaliplatin administration in clinical trials.
Collapse
Affiliation(s)
- Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Ran
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cuihan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Song
- Department of Pharmacy, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Wang Q, Liu P, Wen Y, Li K, Bi B, Li BB, Qiu M, Zhang S, Li Y, Li J, Chen H, Yin Y, Zeng L, Zhang C, He Y, Zhao J. Metal-enriched HSP90 nanoinhibitor overcomes heat resistance in hyperthermic intraperitoneal chemotherapy used for peritoneal metastases. Mol Cancer 2023; 22:95. [PMID: 37316830 DOI: 10.1186/s12943-023-01790-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/16/2023] [Indexed: 06/16/2023] Open
Abstract
Clinical hyperthermic intraperitoneal chemotherapy (HIPEC) is regarded as a potential treatment that can prolong survival of patients with peritoneal metastases after cytoreductive surgery. However, treated tumor cells are prone to becoming heat resistant to HIPEC therapy through high expression of heat shock proteins (HSPs). Here, a carrier-free bifunctional nanoinhibitor was developed for HIPEC therapy in the management of peritoneal metastases. Self-assembly of the nanoinhibitor was formed by mixing Mn ion and epigallocatechin gallate (EGCG) in a controllable manner. Such nanoinhibitor directly inhibited HSP90 and impaired the HSP90 chaperone cycle by reduced intracellular ATP level. Additionally, heat and Mn ion synergistically induced oxidative stress and expression of caspase 1, which activated GSDMD by proteolysis and caused pyroptosis in tumor cells, triggering immunogenic inflammatory cell death and induced maturation of dendritic cells through the release of tumor antigens. This strategy to inhibit heat resistance in HIPEC presented an unprecedented paradigm for converting "cold" tumors into "hot" ones, thus significantly eradicating disseminated tumors located deep in the abdominal cavity and stimulating immune response in peritoneal metastases of a mouse model. Collectively, the nanoinhibitor effectively induced pyroptosis of colon tumor cells under heat conditions by inhibiting heat stress resistance and increasing oxidative stress, which may provide a new strategy for treatment of colorectal peritoneal metastases.
Collapse
Affiliation(s)
- Qiang Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Peng Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Kuan Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Bo Bi
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Bin-Bin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Miaojuan Qiu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jia Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Hengxing Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Yuan Yin
- Gastric Cancer Center, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Sichuan, China
| | - Leli Zeng
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Jing Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China.
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
19
|
Jäger T, Schredl P, Neureiter D, Presl J, Tschann P, Königsrainer I, Pascher A, Emmanuel K, Regenbogen S, Ramspott JP. The SAlzburg PEritoneal SUrface CAlculator (SAPESUCA): The First Web-Based Application for Peritoneal Surface Area Quantification. Cancers (Basel) 2023; 15:3134. [PMID: 37370744 DOI: 10.3390/cancers15123134] [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: 02/16/2023] [Revised: 05/05/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: Peritoneal metastasized colorectal cancer is associated with a worse prognosis. The combination of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) showed promising results in selected patients, but standardization is lacking so far. We present the first tool enabling standardized peritoneal surface area (PSA) quantification in patients undergoing CRS and HIPEC: The SAlzburg PEritoneal SUrface CAlculator (SAPESUCA). (2) Methods: SAPESUCA was programmed using the R-Shiny framework. The application was validated in 23 consecutive colon cancer patients who received 27 closed oxaliplatin-based HIPECs between 2016 and 2020. The programming algorithm incorporates the patient's body surface area and its correlated peritoneal surface area (PSA) based on the 13 Peritoneal Cancer Index (PCI) regions. (3) Results: Patients' median age was 56 years. Median PCI was 9. SAPESUCA revealed a mean PSA of 18,613 cm2 ± 1951 of all patients before compared to 13,681 cm2 ± 2866 after CRS. The Central PCI region revealed the highest mean peritonectomy extent (1517 cm2 ± 737). The peritonectomy extent correlated significantly with PCI score and postoperative morbidity. The simulated mean oxaliplatin dose differed significantly before and after CRS (558 mg/m2 ± 58.4 vs. 409 mg/m2 ± 86.1; p < 0.0001). (4) Conclusion: SAPESUCA is the first free web-based app for standardized determination of the resected and remaining PSA after CRS. The tool enables chemotherapeutic dose adjustment to the remaining PSA.
Collapse
Affiliation(s)
- Tarkan Jäger
- Department of Surgery, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Philipp Schredl
- Department of Surgery, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Jaroslav Presl
- Department of Surgery, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Peter Tschann
- Department of General and Thoracic Surgery, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Ingmar Königsrainer
- Department of General and Thoracic Surgery, Academic Teaching Hospital Feldkirch, 6800 Feldkirch, Austria
| | - Andreas Pascher
- Department for General, Visceral and Transplant Surgery, University Hospital Muenster, 48149 Muenster, Germany
| | - Klaus Emmanuel
- Department of Surgery, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Stephan Regenbogen
- Department of Surgery, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Department for Trauma Surgery, BG Trauma Center Murnau, 82418 Murnau, Germany
- Department for Trauma Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071 Ludwigshafen am Rhein, Germany
| | - Jan Philipp Ramspott
- Department of Surgery, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Department for General, Visceral and Transplant Surgery, University Hospital Muenster, 48149 Muenster, Germany
| |
Collapse
|
20
|
Kepenekian V, Sgarbura O, Marchal F, Villeneuve L, Glehen O, Kusamura S, Deraco M. Peritoneal Mesothelioma: Systematic Review of Hyperthermic Intraperitoneal Chemotherapy (HIPEC) Protocol Outcomes. Indian J Surg Oncol 2023; 14:39-59. [PMID: 37359920 PMCID: PMC10284774 DOI: 10.1007/s13193-023-01728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/25/2023] [Indexed: 03/07/2023] Open
Abstract
Diffuse malignant peritoneal mesothelioma (DMPM) prognosis was improved by the locoregional treatment combining cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). HIPEC is a multiparametric treatment with multiple protocols proposed and reviewed in this work. A systematic review of medical literature was performed according to PRISMA guidelines. The search strategy used "malignant peritoneal mesothelioma" and "HIPEC" as keywords in three databases. Studies were included if reporting precisely the HIPEC regimen and the related outcomes, if comparing regimen, or if reporting national/international guidelines. The GRADE methodology was used to rate the level of evidence. Twenty-eight studies were included in this review: 1 was a meta-analysis, 18 reported cohort outcomes, 4 retrospectively compared HIPEC regimens, and 5 were guidelines. Six HIPEC regimens were found, 4 with one drug (cisplatin, mitomycine-C, carboplatin, oxaliplatin), 2 using two drugs (cisplatin-doxorubicin or cisplatin-mitomycine-C). Cisplatin, up to 250 mg/m2 over 90 min, appeared as the key HIPEC drug with a toxicity profile well controlled by the concomitant intravenous perfusion of sodium thiosulfate. Comparative studies tended to show that a bi-drug regimen led to better long-term oncologic outcomes, with cisplatin 50 mg/m2 plus doxorubicin 15 mg/m2 being safe and more efficient. This late protocol was the most widely used and recommended in 3 out of 4 international guidelines. Cisplatin was the preferred drug for HIPEC in DMPM patients. Most of the time, it was combined with doxorubicin for 90 min. A harmonization of protocols and further comparative studies are needed to optimize HIPEC regimen choice.
Collapse
Affiliation(s)
- Vahan Kepenekian
- Service de Chirurgie Digestive, Hôpital Lyon Sud, Hospices Civils de Lyon, -69495 Pierre-Bénite F, France
- EA 3738 CICLY, Université Lyon-1, -69921 Oullins cedex F, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Université Claude Bernard Lyon 1, CICLY, Lyon, France
- Service de Chirurgie Oncologique, Hôpital Lyon Sud, 165, Chemin du Grand Revoyet, - 69310 Pierre Bénite, France
| | - Olivia Sgarbura
- Surgical Oncology Department, Montpellier Cancer Institute (ICM), University of Montpellier, Montpellier, France
| | - Frederic Marchal
- Department of Surgical Oncology, Institut de Cancérologie de Lorraine, Université de Lorraine, Nancy, France
| | - Laurent Villeneuve
- EA 3738 CICLY, Université Lyon-1, -69921 Oullins cedex F, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Université Claude Bernard Lyon 1, CICLY, Lyon, France
- Service de Recherche Et d’Epidémiologie Cliniques, Hôpital Lyon Sud, Hospices Civils de Lyon, -69495 Pierre-Bénite F, France
| | - Olivier Glehen
- Service de Chirurgie Digestive, Hôpital Lyon Sud, Hospices Civils de Lyon, -69495 Pierre-Bénite F, France
- EA 3738 CICLY, Université Lyon-1, -69921 Oullins cedex F, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Université Claude Bernard Lyon 1, CICLY, Lyon, France
| | - Shigeki Kusamura
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marcello Deraco
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
21
|
Frankinet L, Bhatt A, Alcazer V, Classe JM, Bereder JM, Meeus P, Pomel C, Mithieux F, Abboud K, Wermert R, Lavoue V, Marchal F, Glehen O, Bakrin N. Role of Hyperthermic Intraperitoneal Chemotherapy Combined with Cytoreductive Surgery as Consolidation Therapy for Advanced Epithelial Ovarian Cancer. Ann Surg Oncol 2023; 30:3287-3299. [PMID: 36820940 DOI: 10.1245/s10434-023-13242-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/28/2022] [Indexed: 02/24/2023]
Abstract
BACKGROUND Patients with advanced epithelial ovarian cancer who undergo incomplete surgery followed by six cycles of chemotherapy could benefit from second-look or consolidation cytoreductive surgery (CCRS). The primary goal of this study was to evaluate the overall survival (OS) in patients undergoing complete CCRS and the factors affecting survival. The secondary goal was to study the benefit of hyperthermic intraperitoneal chemotherapy (HIPEC) in these patients. METHODS This was a retrospective analysis of 173 patients with CCRS with (n = 118) or without (n = 55) HIPEC treated at 12 French centers. Only patients having a completeness of cytoreduction (CC) 0/1 resection and a minimum of 5 years of follow-up were included. HIPEC was performed systematically for all patients except those treated at the four centers that did not perform HIPEC. RESULTS The median Peritoneal Cancer Index was 6 (range 0-33). Closed HIPEC was performed in 59 (34.1%) patients and open HIPEC was performed in 56 (32.3%) patients. Grade 3-4 complications occurred in 64 (36.9%) patients. The median OS was 35.67 months (95% confidence interval [CI] 29.8-46.1) and was significantly longer for CCRS + HIPEC (31.4 months without HIPEC and 42.5 months with HIPEC; p = 0.022). On multivariate analysis, closed HIPEC (hazard ratio [HR] 0.46, 95% CI 0.29-0.73; p < 0.001) resulted in a longer OS, and age > 65 years (HR 2.17, 95% CI 1.14-4.11; p = 0.018) and bowel resection (HR 1.98, 95% CI 1.27-3.08; p = 0.020) led to a shorter OS. On multivariate logistic regression analysis, closed HIPEC (odds ratio 0.18; p = 0.001) was associated with a lower risk of dying at 5 years. CONCLUSIONS CCRS was performed with an acceptable morbidity and resulted in good overall survival. The role of HIPEC in addition to CCRS should be evaluated in prospective, randomized studies and the closed technique prospectively compared with the open technique.
Collapse
Affiliation(s)
- Lisa Frankinet
- Department of General Surgery, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Lyon, France
| | - Aditi Bhatt
- Department of Surgical Oncology, Zydus Hospital, Ahmedabad, India
| | - Vincent Alcazer
- Department of Medical Oncology, Centre Hospitalier Lyon Sud, Lyon, France
| | - Jean-Marc Classe
- Department of Surgical Oncology, Centre René Gauducheau, Nantes, France
| | - Jean-Marc Bereder
- Department of Surgical Oncology, Centre Hospitalier L'Archet, Nice, France
| | - Pierre Meeus
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - Christophe Pomel
- Department of Surgical Oncology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Francois Mithieux
- Department of Surgical Oncology, Hopital Privé Jean Mermoz, Lyon, France
| | - Karine Abboud
- Department of Surgical Oncology, Hopital Nord St Etienne, St Étienne, France
| | - Romauld Wermert
- Department of Surgical Oncology, Centre Paul Papin, Angers, France
| | - Vincent Lavoue
- Department of Surgical Oncology, Centre Hospitralo-Universitaire Rennes, Rennes, France
| | - Frederic Marchal
- Department of Surgical Oncology, Institut de Cancérologir de Lorraine Alexis Vautrin, Université de Lorraine, Nancy, France
| | - Olivier Glehen
- Department of General Surgery, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Lyon, France. .,CICLY, Lyon 1 University, Lyon, France.
| | - Naoual Bakrin
- Department of General Surgery, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Lyon, France.,CICLY, Lyon 1 University, Lyon, France
| |
Collapse
|
22
|
Guchelaar NAD, Noordman BJ, Koolen SLW, Mostert B, Madsen EVE, Burger JWA, Brandt-Kerkhof ARM, Creemers GJ, de Hingh IHJT, Luyer M, Bins S, van Meerten E, Lagarde SM, Verhoef C, Wijnhoven BPL, Mathijssen RHJ. Intraperitoneal Chemotherapy for Unresectable Peritoneal Surface Malignancies. Drugs 2023; 83:159-180. [PMID: 36633826 PMCID: PMC9908703 DOI: 10.1007/s40265-022-01828-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
Malignancies of the peritoneal cavity are associated with a dismal prognosis. Systemic chemotherapy is the gold standard for patients with unresectable peritoneal disease, but its intraperitoneal effect is hampered by the peritoneal-plasma barrier. Intraperitoneal chemotherapy, which is administered repeatedly into the peritoneal cavity through a peritoneal implanted port, could provide a novel treatment modality for this patient population. This review provides a systematic overview of intraperitoneal used drugs, the performed clinical studies so far, and the complications of the peritoneal implemental ports. Several anticancer drugs have been studied for intraperitoneal application, with the taxanes paclitaxel and docetaxel as the most commonly used drug. Repeated intraperitoneal chemotherapy, mostly in combination with systemic chemotherapy, has shown promising results in Phase I and Phase II studies for several tumor types, such as gastric cancer, ovarian cancer, colorectal cancer, and pancreatic cancer. Two Phase III studies for intraperitoneal chemotherapy in gastric cancer have been performed so far, but the results regarding the superiority over standard systemic chemotherapy alone, are contradictory. Pressurized intraperitoneal administration, known as PIPAC, is an alternative way of administering intraperitoneal chemotherapy, and the first prospective studies have shown a tolerable safety profile. Although intraperitoneal chemotherapy might be a standard treatment option for patients with unresectable peritoneal disease, more Phase II and Phase III studies focusing on tolerability profiles, survival rates, and quality of life are warranted in order to establish optimal treatment schedules and to establish a potential role for intraperitoneal chemotherapy in the approach to unresectable peritoneal disease.
Collapse
Affiliation(s)
- Niels A. D. Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Bo J. Noordman
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
- Department of Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Eva V. E. Madsen
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Alexandra R. M. Brandt-Kerkhof
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Ignace H. J. T. de Hingh
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Misha Luyer
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Sjoerd M. Lagarde
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Bas P. L. Wijnhoven
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron. H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
23
|
Vaira M, Robella M, Guaglio M, Berchialla P, Sommariva A, Valle M, Pasqual EM, Roviello F, Framarini M, Fiorentini G, Sammartino P, Ilari Civit A, Di Giorgio A, Ansaloni L, Deraco M. Diagnostic and Therapeutic Algorithm for Appendiceal Tumors and Pseudomyxoma Peritonei: A Consensus of the Peritoneal Malignancies Oncoteam of the Italian Society of Surgical Oncology (SICO). Cancers (Basel) 2023; 15:cancers15030728. [PMID: 36765686 PMCID: PMC9913318 DOI: 10.3390/cancers15030728] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/28/2022] [Accepted: 12/07/2022] [Indexed: 01/26/2023] Open
Abstract
Aim: Pseudomyxoma peritonei (PMP) is an uncommon pathology, and its rarity causes a lack of scientific evidence, precluding the design of a prospective trial. A diagnostic and therapeutic algorithm (DTA) is necessary in order to standardize the disease treatment while balancing optimal patient management and the correct use of resources. The Consensus of the Italian Society of Surgical Oncology (SICO) Oncoteam aims at defining a diagnostic and therapeutic pathway for PMP and appendiceal primary tumors applicable in Italian healthcare. Method: The consensus panel included 10 delegated representatives of oncological referral centers for Peritoneal Surface Malignancies (PSM) affiliated to the SICO PSM Oncoteam. A list of statements regarding the DTA of patients with PMP was prepared according to recommendations based on the review of the literature and expert opinion. Results: A consensus was obtained on 33 of the 34 statements linked to the DTA; two flowcharts regarding the management of primary appendiceal cancer and peritoneal disease were approved. Conclusion: Currently, consensus has been reached on pathological classification, preoperative evaluation, cytoreductive surgery technical detail, and systemic treatment; some controversies still exist regarding the exclusion criteria for HIPEC treatment. A shared Italian model of DTA is an essential tool to ensure the appropriateness and equity of treatment for these patients.
Collapse
Affiliation(s)
- Marco Vaira
- Unit of Surgical Oncology, Candiolo Cancer Institute, FPO—IRCCS, 10060 Candiolo, Italy
- Correspondence: authors: (M.V.); (M.R.)
| | - Manuela Robella
- Unit of Surgical Oncology, Candiolo Cancer Institute, FPO—IRCCS, 10060 Candiolo, Italy
- Correspondence: authors: (M.V.); (M.R.)
| | - Marcello Guaglio
- Peritoneal Surface Malignancies Unit, Fondazione Istituto Nazionale Tumori IRCCS Milano, 20133 Milano, Italy
| | - Paola Berchialla
- Department of Clinical and Biological Sciences, Centre for Biostatistics, Epidemiology and Public Health (C-BEPH), University of Torino, 10124 Torino, Italy
| | - Antonio Sommariva
- Advanced Surgical Oncology Unit, Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, t, 35100 Padova, Italy
| | - Mario Valle
- Peritoneal Tumours Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Enrico Maria Pasqual
- AOUD Center Advanced Surgical Oncology, DAME University of Udine, 33100 Udine, Italy
| | - Franco Roviello
- Unit of General Surgery and Surgical Oncology, Department of Medicine, Surgery, and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Massimo Framarini
- Surgery and Advanced Oncological Therapy Unit, Ospedale “GB.Morgagni-L.Pierantoni”—AUSL Forlì, 47122 Forlì, Italy
| | - Giammaria Fiorentini
- Italian Network of International Clinical Hyperthermia Society Coordinator, 48121 Ravenna, Italy
| | - Paolo Sammartino
- CRS and HIPEC Unit, Pietro Valdoni, Umberto I Policlinico di Roma, 00161 Roma, Italy
| | - Alba Ilari Civit
- Unit of Surgical Oncology, Candiolo Cancer Institute, FPO—IRCCS, 10060 Candiolo, Italy
| | - Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli—IRCCS, 00168 Rome, Italy
| | - Luca Ansaloni
- Unit of General Surgery, San Matteo Hospital, 27100 Pavia, Italy
| | - Marcello Deraco
- Peritoneal Surface Malignancies Unit, Fondazione Istituto Nazionale Tumori IRCCS Milano, 20133 Milano, Italy
| |
Collapse
|
24
|
Foster JM, Zhang C, Rehman S, Sharma P, Alexander HR. The contemporary management of peritoneal metastasis: A journey from the cold past of treatment futility to a warm present and a bright future. CA Cancer J Clin 2023; 73:49-71. [PMID: 35969103 DOI: 10.3322/caac.21749] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/12/2022] [Accepted: 06/15/2022] [Indexed: 01/17/2023] Open
Abstract
Peritoneal metastasis (PM) is often regarded as a less frequent pattern of spread; however, collectively across all spectra of primary tumors, the consequences of PM impact a large population of patients annually. Unlike other modes of metastasis, symptoms at presentation or during the treatment course are common, representing an additional challenge in the management of PM. Early efforts with chemotherapy and incomplete surgical interventions transiently improved symptoms, but durable symptom control and survival extension were rare, which established a perspective of treatment futility for PM through most of the 20th century. Notably, the continued development of better systemic therapy combinations, optimization of cytoreductive surgery (CRS), and rigorous investigation of combining regional therapy-specifically hyperthermic intraperitoneal chemotherapy-with CRS, have resulted in more effective multimodal treatment options for patients with PM. In this article, the authors provide a comprehensive review of the data establishing the contemporary approach for tumors with a high frequency of PM, including appendix, colorectal, mesothelioma, and gastric cancers. The authors also explore the emerging role of adding hyperthermic intraperitoneal chemotherapy to the well established paradigm of CRS and systemic therapy for advanced ovarian cancer, as well as the recent clinical trials identifying the efficacy of poly(adenosine diphosphate ribose) polymerase maintenance therapy. Finally, recent data are included that explore the role of precision medicine technology in PM management that, in the future, may help further improve patient selection, identify the best systemic therapy regimens, detect actionable mutations, and identify new targets for drug development.
Collapse
Affiliation(s)
- Jason M Foster
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chunmeng Zhang
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shahyan Rehman
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey
| | - Prateek Sharma
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | |
Collapse
|
25
|
Vandaele T, Van Slambrouck J, Proesmans V, Clement P, Lambrecht M, Nafteux P, Van Raemdonck D, Ceulemans LJ. Hyperthermic Intrathoracic Chemotherapy (HITHOC) for Pleural Disseminated Thymoma: A Systematic Literature Review. Ann Surg Oncol 2023; 30:543-560. [PMID: 36151429 DOI: 10.1245/s10434-022-12461-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Optimal treatment for thymoma with pleural dissemination (TPD) remains unclear. Extended radical resection is the cornerstone for local treatment but the need for pleuro-pneumonectomy is debatable. Cytoreductive surgery with intraoperative hyperthermic intrathoracic chemotherapy (HITHOC) provides an alternative strategy to reduce tumor load and prevent pleural recurrence. OBJECTIVE The aim of this review was to provide an overview of current literature regarding HITHOC for TPD. METHODS A systematic literature review (PRISMA) was performed in the EMBASE, MEDLINE, Cochrane and Web of Science databases, resulting in 154 papers selected for screening (PROSPERO: CRD42020208242). Title, abstract, and full-text screening resulted in 13 papers subjected to structured data extraction and methodological quality assessment. One additional case from our department was included. Inclusion criteria were original research reporting on patients diagnosed with TPD; oncological outcome reporting; intraoperative HITHOC; and papers written in English, Dutch or German. Methodological quality was assessed using the Risk-of-Bias (RoB)-2 Tool and the Newcastle-Ottawa scale. RESULTS HITHOC for TPD was reported in 171 cases. HITHOC-related mortality was absent and morbidity was reported in three cases. Intrathoracic perfusion of a platinum-derivative, often combined with other chemotherapeutic drugs at >40°C for 60 min or longer was always used. Post-HITHOC recurrence was reported in 37/120 cases (31%). In patients with a minimal 1-year follow-up, average time to recurrence was 68.5 months. CONCLUSION Combining cytoreductive surgery and HITHOC is feasible and safe for TPD. The strong heterogeneity in the literature impedes proper outcome analysis. More research is needed to better understand the additional benefit of HITHOC in the TPD setting.
Collapse
Affiliation(s)
- Tom Vandaele
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Viktor Proesmans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Paul Clement
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium.,Laboratory of Experimental Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Lambrecht
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium.,Laboratory of Experimental Radiotherapy, University Hospitals Leuven, Leuven, Belgium
| | - Philippe Nafteux
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Dirk Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium. .,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.
| |
Collapse
|
26
|
Menzies AV, Usher EC, Hsu FC, Levine EA, Lentz SS, Kelly MG. HIPEC after neoadjuvant chemotherapy is associated with acceptable toxicity and favorable quality of life in newly diagnosed advanced ovarian cancer patients. Gynecol Oncol 2022; 167:234-238. [PMID: 36085091 DOI: 10.1016/j.ygyno.2022.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To evaluate toxicity, quality of life and PFS in patients with advanced ovarian cancer who underwent neoadjuvant chemotherapy (NAC) followed by CRS and HIPEC with carboplatin. METHODS Patients with stage IIIC or IVA epithelial ovarian cancer, who were not candidates for primary CRS, were enrolled in this phase two trial. Patients received 3-6 cycles of NAC with an IV carboplatin doublet followed by CRS with HIPEC (carboplatin 800 mg/m2 for 90 min). They were followed for at least 12 months to assess for adverse events, quality of life (QOL) and disease progression. QOL was measured using the Functional Assessment of Cancer Therapy-Ovarian (FACT-O) questionnaires prior to CRS and post-operatively at 6 weeks, 3 months, and 6 months after CRS. RESULTS Twenty patients were enrolled. HIPEC was completed successfully in all twenty patients, and there was no peri-operative mortality. Twelve (70.6%) patients experienced a grade 3 or 4 toxicity; most commonly anemia (59%), thrombocytopenia (29%), and hypokalemia (24%). There was no significant change between the pre-operative and postoperative 6 weeks, 3 month, and 6 month FACT-O, NTX, and AD scores. Nine (45%) patients have experienced disease recurrence to date. The median progression free survival in this cohort is 11.2 months (2.5-23.7 months). CONCLUSION The addition of HIPEC with carboplatin to interval CRS was well tolerated in patient population. Myelosuppression was the most common adverse event. CRS with HIPEC did not adversely impact these patients' QOL indices. The efficacy of this regimen should be further evaluated in a larger clinical trial.
Collapse
Affiliation(s)
- Anya V Menzies
- Department of Obstetrics and Gynecology, Atrium Health Wake Forest Baptist, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America.
| | - Erik C Usher
- Department of Obstetrics and Gynecology, Atrium Health Wake Forest Baptist, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America
| | - Edward A Levine
- Department of Surgical Oncology, Atrium Health Wake Forest Baptist, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America
| | - Samuel S Lentz
- Department of Obstetrics and Gynecology, Atrium Health Wake Forest Baptist, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America
| | - Michael G Kelly
- Department of Obstetrics and Gynecology, Atrium Health Wake Forest Baptist, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America
| |
Collapse
|
27
|
Proesmans V, Vandaele T, Van Slambrouck J, Wolthuis A, D Hoore A, Dekervel J, Van Cutsem E, Dresen R, De Hertogh G, Degezelle K, Van Raemdonck D, Nafteux P, Ceulemans LJ. Pleural decortication and hyperthermic intrathoracic chemotherapy for pseudomyxoma. Int J Hyperthermia 2022; 39:1153-1157. [PMID: 36049823 DOI: 10.1080/02656736.2022.2104938] [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] [Indexed: 10/14/2022] Open
Abstract
Aim: Pleural dissemination of pseudomyxoma peritonei (PMP) is an extremely rare diagnosis, for which no standard therapy is available.Methods: We describe the successful treatment of a 67-year-old male diagnosed with left-sided intrapleural dissemination of PMP (low-grade appendiceal mucinous neoplasm), 2 years after treatment of abdominal PMP with cytoreductive surgery (CRS) and hyperthermic intra-peritoneal chemotherapy. Treatment consisted of extended pleural decortication (ePD) and oxaliplatin-based hyperthermic intrathoracic chemotherapy (HITHOC). The patient is doing well without complications or signs of recurrence, 26 months after thoracic surgery.Conclusion: ePD in combination with HITHOC is a valuable treatment for thoracic PMP.
Collapse
Affiliation(s)
- Viktor Proesmans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Tom Vandaele
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Albert Wolthuis
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Oncology, laboratory of Abdominal Surgical Oncology, KU Leuven, Leuven, Belgium
| | - André D Hoore
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Oncology, laboratory of Abdominal Surgical Oncology, KU Leuven, Leuven, Belgium
| | - Jeroen Dekervel
- Department of Gastro-Enterology, University Hospitals Leuven, Leuven, Belgium.,Department of Oncology, Laboratory of Clinical Digestive Oncology, KU Leuven, Leuven, Belgium
| | - Eric Van Cutsem
- Department of Gastro-Enterology, University Hospitals Leuven, Leuven, Belgium.,Department of Oncology, Laboratory of Clinical Digestive Oncology, KU Leuven, Leuven, Belgium
| | - Raphaëla Dresen
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium.,Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Gert De Hertogh
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Karlien Degezelle
- Department of Cardiac Surgery, Division Perfusion, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Philippe Nafteux
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Park EJ, Ahn J, Abuzar SM, Park KS, Hwang SJ, Baik SH. Pharmacologic Effects of Oxaliplatin Instability in Chloride-Containing Carrier Fluids on the Hyperthermic Intraperitoneal Chemotherapy to Treat Colorectal Cancer In Vitro and In Vivo. Ann Surg Oncol 2022; 29:8583-8592. [DOI: 10.1245/s10434-022-12358-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/12/2022] [Indexed: 01/05/2023]
|
29
|
Strach MC, Sutherland S, Horvath LG, Mahon K. The role of chemotherapy in the treatment of advanced appendiceal cancers: summary of the literature and future directions. Ther Adv Med Oncol 2022; 14:17588359221112478. [PMID: 35898968 PMCID: PMC9310237 DOI: 10.1177/17588359221112478] [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: 01/08/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Appendiceal cancer is rare and encompasses a diverse group of tumours ranging from low-grade appendiceal mucinous neoplasms to high-grade adenocarcinomas. Appendiceal cancers often spread to the peritoneal cavity causing extensive mucinous dissemination and peritoneal metastases. Prognosis varies with histological subtype. Cytoreductive surgery and heated intraperitoneal chemotherapy is well-established as the most effective treatment achieving long-term survival in some patients. Chemotherapy regimens used to treat appendiceal cancer are extrapolated from the colorectal cancer setting, but disease biology differs and outcomes are inferior. The role of chemotherapy in the treatment of appendiceal cancer remains poorly defined. There is an urgent need to develop novel tailored treatment strategies in the perioperative and unresectable setting. This review aims to evaluate the literature for patients who received intraperitoneal and systemic chemotherapy for appendiceal cancers.
Collapse
Affiliation(s)
| | | | | | - Kate Mahon
- Chris O'Brien Lifehouse, 119-144 Missenden Road, Camperdown, NSW 2050, Australia
| |
Collapse
|
30
|
Jafari MD, Carmichael JC, Dayyani F, McKinney C, Wenzel L, Zell JA, Pigazzi A. Immediate Adjuvant Chemotherapy in Non-Metastatic Colon Cancer: Phase I Trial Evaluating a Novel Treatment Protocol. Clin Colorectal Cancer 2022; 21:114-121. [PMID: 34980534 PMCID: PMC9844669 DOI: 10.1016/j.clcc.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/19/2021] [Accepted: 11/22/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND The optimal timing of adjuvant chemotherapy (AC) in non-metastatic colon cancer is poorly defined. Delays in AC result in decreased survival. Effective cytotoxic treatments should be considered during the perioperative phase of care. The immediate adjuvant chemotherapy (IAC) concept intends to capitalize on the therapeutic benefits that can be achieved in the perioperative period. We aim to demonstrate that IAC is safe and tolerable. PATIENT AND METHODS Microsatellite stable invasive adenocarcinomas were treated with intravenous Leucovorin 20 mg/m2 and single dose of 5-Flurouracil 400mg/m2 at the time of surgery. High-risk stage II and stage III received the first dose of standard AC at 14 days after surgery. Serial measurements of blood-based biomarkers were measured. Quality of life (QOL) was measured using EORTC QLQ-C30. RESULTS Of the 20 patients recruited, 40% had final pathology of stage III, 40% stage II and 20% stage I. All patients received intra-operative chemotherapy with no associated morbidity. Median length of stay was 2 days (range of 2-4). There was no intraoperative morbidity with 5% (N = 1) grade 3 complication. AC was administered to 65% of patients. The median time to AC was 14 days (range 14-36). Overall quality of life and health scores were similar before surgery and at 30-day postoperatively (P < .05). CONCLUSIONS A protocol based on IAC starting at the time of surgical resection was found to be safe and feasible with no adverse effects on surgical morbidity or quality of life. Further prospective studies are needed to explore the oncologic benefit of this novel systemic treatment approach.
Collapse
Affiliation(s)
| | - Joseph C. Carmichael
- Division of Colorectal Surgery, Department of Surgery, University of California,Irvine Medical Center, CA
| | - Farshid Dayyani
- Department of Medicine, Division of Oncology, University of California, Irvine Medical Center, CA
| | - Chelsea McKinney
- Department of Medicine, Division of Oncology, University of California, Irvine Medical Center, CA
| | - Lari Wenzel
- Department of Medicine, Division of Oncology, University of California, Irvine Medical Center, CA
| | - Jason A. Zell
- Department of Medicine, Division of Oncology, University of California, Irvine Medical Center, CA
| | | |
Collapse
|
31
|
Current Trends in Cytoreductive Surgery (CRS) and Hyperthermic Intraperitoneal Chemotherapy (HIPEC) for Peritoneal Disease from Appendiceal and Colorectal Malignancies. J Clin Med 2022; 11:jcm11102840. [PMID: 35628966 PMCID: PMC9143396 DOI: 10.3390/jcm11102840] [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: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/10/2022] Open
Abstract
Peritoneal carcinomatosis (PC) is a poor prognostic factor for all malignancies. This extent of metastatic disease progression remains difficult to treat with systemic therapies due to poor peritoneal vascularization resulting in limited drug delivery and penetration into tissues. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are surgical interventions that directly target peritoneal tumors and have improved outcomes for PC resulting from appendiceal and colorectal cancer (CRC). Despite these radical therapies, long-term survival remains infrequent, and recurrence is common. The reasons for these outcomes are multifactorial and signal the need for the continued development of novel therapeutics, techniques, and approaches to improve outcomes for these patients. Here, we review landmark historical studies that serve as the foundation for current recommendations, recent discoveries, clinical trials, active research, and areas of future interest in CRS/HIPEC to treat PC originating from appendiceal and colorectal malignancies.
Collapse
|
32
|
Pereira F, Serrano A, Manzanedo I, Pérez-Viejo E, González-Moreno S, González-Bayón L, Arjona-Sánchez A, Torres J, Ramos I, Barrios ME, Cascales P, Morales R, Boldó E, García-Fadrique A, Arteaga X, Gutierrez-Calvo A, Sánchez-García S, Asensio E, Ramírez CP, Artiles M, Vaqué J, Parra PA, Villarejo P, Muñoz-Casares C, Turienzo E, Calero A, Torrejimeno IJ, Prieto I, Galindo J, Borrego V, Marcello ME, Rihuete C, Carrasco J, Gomez-Quiles L. GECOP-MMC: phase IV randomized clinical trial to evaluate the efficacy of hyperthermic intraperitoneal chemotherapy (HIPEC) with mytomicin-C after complete surgical cytoreduction in patients with colon cancer peritoneal metastases. BMC Cancer 2022; 22:536. [PMID: 35549912 PMCID: PMC9097342 DOI: 10.1186/s12885-022-09572-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/19/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The French PRODIGE 7 trial, published on January 2021, has raised doubts about the specific survival benefit provided by HIPEC with oxaliplatin 460 mg/m2 (30 minutes) for the treatment of peritoneal metastases from colorectal cancer. However, several methodological flaws have been identified in PRODIGE 7, specially the HIPEC protocol or the choice of overall survival as the main endpoint, so its results have not been assumed as definitive, emphasizing the need for further research on HIPEC. It seems that the HIPEC protocol with high-dose mytomicin-C (35 mg/m2) is the preferred regime to evaluate in future clinical studies. METHODS GECOP-MMC is a prospective, open-label, randomized, multicenter phase IV clinical trial that aims to evaluate the effectiveness of HIPEC with high-dose mytomicin-C in preventing the development of peritoneal recurrence in patients with limited peritoneal metastasis from colon cancer (not rectal), after complete surgical cytoreduction. This study will be performed in 31 Spanish HIPEC centres, starting in March 2022. Additional international recruiting centres are under consideration. Two hundred sixteen patients with PCI ≤ 20, in which complete cytoreduction (CCS 0) has been obtained, will be randomized intraoperatively to arm 1 (with HIPEC) or arm 2 (without HIPEC). We will stratified randomization by surgical PCI (1-10; 11-15; 16-20). Patients in both arms will be treated with personalized systemic chemotherapy. Primary endpoint is peritoneal recurrence-free survival at 3 years. An ancillary study will evaluate the correlation between surgical and pathological PCI, comparing their respective prognostic values. DISCUSSION HIPEC with high-dose mytomicin-C, in patients with limited (PCI ≤ 20) and completely resected (CCS 0) peritoneal metastases, is assumed to reduce the expected risk of peritoneal recurrence from 50 to 30% at 3 years. TRIAL REGISTRATION EudraCT number: 2019-004679-37; Clinicaltrials.gov: NCT05250648 (registration date 02/22/2022, ).
Collapse
Affiliation(s)
- Fernando Pereira
- Hospital Universitario de Fuenlabrada, Camino del Molino 2, Fuenlabrada, 28942, Madrid, Spain.
| | - Angel Serrano
- Hospital Universitario de Fuenlabrada, Camino del Molino 2, Fuenlabrada, 28942, Madrid, Spain
| | - Israel Manzanedo
- Hospital Universitario de Fuenlabrada, Camino del Molino 2, Fuenlabrada, 28942, Madrid, Spain
| | - Estibalitz Pérez-Viejo
- Hospital Universitario de Fuenlabrada, Camino del Molino 2, Fuenlabrada, 28942, Madrid, Spain
| | | | - Luis González-Bayón
- Hospital General Universitario Gregorio Marañón, C/ Doctor Esquerdo, 46 -, 28007, Madrid, Spain
| | | | - Juan Torres
- Hospital Universitario Torrecárdenas, Calle Hermandad de Donantes de Sangre s/n, 04009, Almería, Spain
| | - Isabel Ramos
- Hospital Sant Joan Despi Moises Broggi, Carrer de Jacint Verdaguer, 90, 08970 Sant Joan Despí, Barcelona, Spain
| | - Maria E Barrios
- Hospital Clinico Universitario de Valencia, Avenida de Blasco Ibañez, 17, 46010, Valencia, Spain
| | - Pedro Cascales
- Hospital Universitario Virgen de la Arrixaca, Ctra. Madrid-Cartagena, s/n, 30120, El Palmar, Murcia, Spain
| | - Rafael Morales
- Hospital Universitario Son Espases, Carretera de Valldemossa, 79. 07210 Palma, Mallorca, Spain
| | - Enrique Boldó
- Consorcio Hospitalario Provincial De Castellón, Avenida del Doctor Clarà 19, 12006, Castellón de la Plana, Spain
| | | | - Xabier Arteaga
- Hospital Universitario Donostia, Begiristain Doktorea Pasealekua 109, 20014, Donostia, Gipuzkoa, Spain
| | - Alberto Gutierrez-Calvo
- Hospital Universitario Principe de Asturias, Carretera Alcalá-Meco, s/n - 28805 Alcalá de Henares, Madrid, Spain
| | - Susana Sánchez-García
- Hospital General Universitario de Ciudad Real, C/ Obispo Rafael Torija s/n - Pol. Larache, 13005, Ciudad Real, Spain
| | - Enrique Asensio
- Hospital Universitario Río Hortega, Calle Dulzaina, 2, 47012, Valladolid, Spain
| | - Cesar P Ramírez
- Hospital Quirónsalud Málaga, Avenida de Imperio Argentina, 1, 29004, Málaga, Spain
| | - Manuel Artiles
- Hospital Universitario de Gran Canaria Doctor Negrín, Barranco de la Ballena, 0, 35010, Las Palmas de Gran Canaria, Spain
| | - Javier Vaqué
- Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Pedro A Parra
- Hospital General Universitario Reina Sofía, Avda. Intendente Jorge Palacios, 1, 30003, Murcia, Spain
| | - Pedro Villarejo
- Fundación Jiménez Díaz, Avda. Reyes Católicos 2, 28040, Madrid, Spain
| | | | - Estrella Turienzo
- Hospital Universitario Central de Asturias, Avenida de Roma, 0, 33011, Oviedo, Spain
| | - Alicia Calero
- Hospital General Universitario de Elche, Camí de la Almazara, 11, 03203 Elche, Alicante, Spain
| | | | - Isabel Prieto
- Hospital Universitario La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Julio Galindo
- Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9,100, 28034, Madrid, Spain
| | - Vicente Borrego
- Hospital Clínico Universitario "Lozano Blesa", Avda. San Juan Bosco, 15, 50009, Zaragoza, Spain
| | - Manuel E Marcello
- Hospital Universitario Fundación Alcorcón, Calle de Budapest, 1, 28922 Alcorcón, Madrid, Spain
| | - Cristina Rihuete
- Hospital Universitario Infanta Elena, Avenida de los Reyes Católicos 21, 28340 Valdemoro, Madrid, Spain
| | - Joaquin Carrasco
- Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82-88, 29010, Málaga, Spain
| | - Luis Gomez-Quiles
- Hospital General Universitario De Castellón, Avenida de Benicassim, 128, 12004, Castellón, Spain
| |
Collapse
|
33
|
Seyfried N, Yurttas C, Burkard M, Oswald B, Tolios A, Herster F, Kauer J, Jäger T, Königsrainer I, Thiel K, Quante M, Rammensee HG, Venturelli S, Schwab M, Königsrainer A, Beckert S, Löffler MW. Prolonged Exposure to Oxaliplatin during HIPEC Improves Effectiveness in a Preclinical Micrometastasis Model. Cancers (Basel) 2022; 14:cancers14051158. [PMID: 35267468 PMCID: PMC8909393 DOI: 10.3390/cancers14051158] [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: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/12/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Absence of survival benefits when adding hyperthermic intraperitoneal chemotherapy (HIPEC) with oxaliplatin to cytoreductive surgery in peritoneal metastasis from colorectal cancer has recently been shown in the randomized controlled PRODIGE 7 trial. We therefore aimed to investigate the effects of this treatment modality in a preclinical micrometastasis model. Cancer cells were incubated with either patient samples obtained during HIPEC procedures or with defined oxaliplatin-containing solutions prepared according to clinically established HIPEC protocols. Our results demonstrate a limited effectiveness of short-term HIPEC in simulations with oxaliplatin to eliminate micrometastases, although we used platinum-sensitive cell lines for our model. Since these results are in line with findings from current research, our studies might offer further convincing evidence and potential explanations for HIPEC futility observed in clinical application. Abstract Cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) was considered a promising treatment for patients with peritoneal metastasis from colorectal cancer. However, the recently published randomized controlled PRODIGE 7 trial failed to demonstrate survival benefits through the addition of short-term oxaliplatin-based HIPEC. Constituting a complex multifactorial treatment, we investigated HIPEC in a preclinical model concerning the elimination of minimal tumor residues, thereby aiming to better understand the size of effects and respective clinical trial results. Patient samples of peritoneal perfusates obtained during HIPEC treatments and oxaliplatin-containing solutions at clinically relevant dosages, conforming with established HIPEC protocols, were assessed regarding their ability to eliminate modelled ~100 µm thickness cancer cell layers. Impedance-based real-time cell analysis and classical end-point assays were used. Flow cytometry was employed to determine the effect of different HIPEC drug solvents on tumor cell properties. Effectiveness of peritoneal perfusate patient samples and defined oxaliplatin-containing solutions proved limited but reproducible. HIPEC simulations for 30 min reduced the normalized cell index below 50% with peritoneal perfusates from merely 3 out of 9 patients within 72 h, indicating full-thickness cytotoxic effects. Instead, prolonging HIPEC to 1 h enhanced these effects and comprised 7 patients’ samples, while continuous drug exposure invariably resulted in complete cell death. Further, frequently used drug diluents caused approximately 25% cell size reduction within 30 min. Prolonging oxaliplatin exposure improved effectiveness of HIPEC to eliminate micrometastases in our preclinical model. Accordingly, insufficient penetration depth, short exposure time, and the physicochemical impact of drug solvents may constitute critical factors.
Collapse
Affiliation(s)
- Nick Seyfried
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; (B.O.); (F.H.); (J.K.); (H.-G.R.)
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, 81675 Munich, Germany
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
- Correspondence:
| | - Markus Burkard
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany; (M.B.); (S.V.)
| | - Benedikt Oswald
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; (B.O.); (F.H.); (J.K.); (H.-G.R.)
| | - Alexander Tolios
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria;
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstraße 17A, 1090 Vienna, Austria
- Center for Medical Statistics, Informatics and Intelligent Systems, Institute of Artificial Intelligence, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Franziska Herster
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; (B.O.); (F.H.); (J.K.); (H.-G.R.)
- Robert Bosch Center for Tumor Diseases (RBCT), Robert Bosch Hospital, Auerbachstr. 110, 70376 Stuttgart, Germany
| | - Joseph Kauer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; (B.O.); (F.H.); (J.K.); (H.-G.R.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany;
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- Department of Hematology, Oncology, and Rheumatology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tarkan Jäger
- Department of Surgery, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria;
| | - Ingmar Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
- Department of General, Visceral and Thoracic Surgery, Landeskrankenhaus Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria
| | - Karolin Thiel
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
| | - Markus Quante
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
| | - Hans-Georg Rammensee
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; (B.O.); (F.H.); (J.K.); (H.-G.R.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tübingen, 72076 Tübingen, Germany
| | - Sascha Venturelli
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany; (M.B.); (S.V.)
- Department of Vegetative and Clinical Physiology, Institute of Physiology, University of Tübingen, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Matthias Schwab
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tübingen, 72076 Tübingen, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, Germany
- Departments of Pharmacy and Biochemistry, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tübingen, 72076 Tübingen, Germany
| | - Stefan Beckert
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
- Department of General and Visceral Surgery, Schwarzwald-Baar Hospital, Klinikstr. 11, 78052 Villingen-Schwenningen, Germany
| | - Markus W. Löffler
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; (N.S.); (I.K.); (K.T.); (M.Q.); (A.K.); (S.B.); (M.W.L.)
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; (B.O.); (F.H.); (J.K.); (H.-G.R.)
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tübingen, 72076 Tübingen, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| |
Collapse
|
34
|
Lurvink RJ, Rovers KP, Wassenaar ECE, Bakkers C, Burger JWA, Creemers GJM, Los M, Mols F, Wiezer MJ, Nienhuijs SW, Boerma D, de Hingh IHJT. Patient-reported outcomes during repetitive oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy for isolated unresectable colorectal peritoneal metastases in a multicenter, single-arm, phase 2 trial (CRC-PIPAC). Surg Endosc 2022; 36:4486-4498. [PMID: 34757489 PMCID: PMC9085665 DOI: 10.1007/s00464-021-08802-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/17/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND CRC-PIPAC prospectively assessed repetitive oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy (PIPAC-OX) as a palliative monotherapy (i.e., without concomitant systemic therapy in between subsequent procedures) for unresectable colorectal peritoneal metastases (CPM). The present study explored patient-reported outcomes (PROs) during trial treatment. METHODS In this single-arm phase 2 trial in two tertiary centers, patients with isolated unresectable CPM received 6-weekly PIPAC-OX (92 mg/m2). PROs (calculated from EQ-5D-5L, and EORTC QLQ-C30 and QLQ-CR29) were compared between baseline and 1 and 4 weeks after the first three procedures using linear mixed modeling with determination of clinical relevance (Cohen's D ≥ 0.50) of statistically significant differences. RESULTS Twenty patients underwent 59 procedures (median 3 [range 1-6]). Several PROs solely worsened 1 week after the first procedure (index value - 0.10, p < 0.001; physical functioning - 20, p < 0.001; role functioning - 27, p < 0.001; social functioning - 18, p < 0.001; C30 summary score - 16, p < 0.001; appetite loss + 15, p = 0.007; diarrhea + 15, p = 0.002; urinary frequency + 13, p = 0.004; flatulence + 13, p = 0.001). These PROs returned to baseline at subsequent time points. Other PROs worsened 1 week after the first procedure (fatigue + 23, p < 0.001; pain + 29, p < 0.001; abdominal pain + 32, p < 0.001), second procedure (fatigue + 20, p < 0.001; pain + 21, p < 0.001; abdominal pain + 20, p = 0.002), and third procedure (pain + 22, p < 0.001; abdominal pain + 22, p = 0.002). Except for appetite loss, all changes were clinically relevant. All analyzed PROs returned to baseline 4 weeks after the third procedure. CONCLUSIONS Patients receiving repetitive PIPAC-OX monotherapy for unresectable CPM had clinically relevant but reversible worsening of several PROs, mainly 1 week after the first procedure. TRIAL REGISTRATION Clinicaltrials.gov: NCT03246321; Netherlands trial register: NL6426.
Collapse
Affiliation(s)
- Robin J. Lurvink
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands ,Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands
| | - Koen P. Rovers
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands
| | - Emma C. E. Wassenaar
- Department of Surgery, St. Antonius Hospital, PO Box 2500, 3430 EM Nieuwegein, The Netherlands
| | - Checca Bakkers
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands
| | - Jacobus W. A. Burger
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands
| | - Geert-Jan M. Creemers
- Department of Medical Oncology, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, St. Antonius Hospital, PO Box 2500, 3430 EM Nieuwegein, The Netherlands
| | - Floortje Mols
- Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands ,Center of Research on Psychology in Somatic Disorders, Department of Medical and Clinical Psychology, Tilburg University, PO Box 90153, 5000 LE Tilburg, The Netherlands
| | - Marinus J. Wiezer
- Department of Surgery, St. Antonius Hospital, PO Box 2500, 3430 EM Nieuwegein, The Netherlands
| | - Simon W. Nienhuijs
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, PO Box 2500, 3430 EM Nieuwegein, The Netherlands
| | - Ignace H. J. T. de Hingh
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands ,Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands ,GROW - School for Oncology and Developmental Biology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
35
|
Alhumaid M, Sait S, Fallatah E, AlSayegh N, Farsi A, Nassif M, Farsi NJ, Akeel N, Samkari A, Shabkah AA, Trabulsi N. Outcomes of Cytoreduction and Oxaliplatin-Based Hyperthermic Intraperitoneal Chemotherapy in Patients With Peritoneal Carcinomatosis From Colorectal Cancer. Cureus 2021; 13:e18670. [PMID: 34786251 PMCID: PMC8579872 DOI: 10.7759/cureus.18670] [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] [Accepted: 10/11/2021] [Indexed: 12/04/2022] Open
Abstract
Among patients with metastatic colorectal cancer, 25% have isolated peritoneal carcinomatosis. We performed a systematic review and meta-analysis to assess the disease-free survival (DFS) and overall survival (OS) of patients undergoing hyperthermic intraperitoneal chemotherapy with oxaliplatin. Eleven studies were included in the final assessment. Pooled three- and five-year OS rates were 58.60% and 42.19%, respectively. The estimated pooled three- and five-year DFS rates were 23.47% and 14.26%, respectively.
Collapse
Affiliation(s)
- Moayad Alhumaid
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Salma Sait
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Emad Fallatah
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Nasser AlSayegh
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Ali Farsi
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Mohammed Nassif
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Nada J Farsi
- Department of Dental Public Health, King Abdulaziz University Faculty of Dentistry, Jeddah, SAU
| | - Nouf Akeel
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Ali Samkari
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Alaa A Shabkah
- Department of Surgery, International Medical Center, Jeddah, SAU
| | - Nora Trabulsi
- Department of Surgery, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| |
Collapse
|
36
|
Zubair M, Adams MS, Diederich CJ. Deployable ultrasound applicators for endoluminal delivery of volumetric hyperthermia. Int J Hyperthermia 2021; 38:1188-1204. [PMID: 34376103 DOI: 10.1080/02656736.2021.1936216] [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: 10/20/2022] Open
Abstract
PURPOSE To investigate the design of an endoluminal deployable ultrasound applicator for delivering volumetric hyperthermia to deep tissue sites as a possible adjunct to radiation and chemotherapy. METHOD This study considers an ultrasound applicator consisting of two tubular transducers situated at the end of a catheter assembly, encased within a distensible conical shaped balloon-based reflector that redirects acoustic energy distally into the tissue. The applicator assembly can be inserted endoluminally or laparoscopically in a compact form and expanded after delivery to the target site. Comprehensive acoustic and biothermal simulations and parametric studies were employed in generalized 3D and patient-specific pancreatic head and body tumor models to characterize the acoustic performance and evaluate heating capabilities of the applicator by investigating the device at a range of operating frequencies, tissue acoustic and thermal properties, transducer configurations, power modulation, applicator positioning, and by analyzing the resultant 40, 41, and 43 °C isothermal volumes and penetration depth of the heating volume. Intensity distributions and volumetric temperature contours were calculated to define moderate hyperthermia boundaries. RESULTS Parametric studies demonstrated the frequency selection to control volume and depth of therapeutic heating from 62 to 22 cm3 and 4 to 2.6 cm as frequency ranges from 1 MHz to 4.7 MHz, respectively. Width of the heating profile tracks closely with the aperture. Water cooling within the reflector balloon was effective in controlling temperature to 37 °C maximum within the luminal wall. Patient-specific studies indicated that applicators with extended OD in the range of 3.6-6.2 cm with 0.5-1 cm long and 1 cm OD transducers can heat volumes of 1.1-7 cm3, 3-26 cm3, and 3.3-37.4 cm3 of pancreatic body and head tumors above 43, 41, and 40 °C, respectively. CONCLUSION In silico studies demonstrated the feasibility of combining endoluminal ultrasound with an integrated expandable balloon reflector for delivering volumetric hyperthermia in regions adjacent to body lumens and cavities.
Collapse
Affiliation(s)
- Muhammad Zubair
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew S Adams
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Chris J Diederich
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
37
|
Sgarbura O, Al Hosni M, Petruzziello A, Figueroa R, Khellaf L, Pissas MH, Carrère S, Nougaret S, Bibeau F, Quénet F. Complete pathologic response after two-stage cytoreductive surgery with HIPEC for bulky pseudomyxoma peritonei: proof of concept. Int J Hyperthermia 2021; 37:585-591. [PMID: 32484014 DOI: 10.1080/02656736.2020.1772511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Introduction: Pseudomyxoma peritonei (PMP) is a rare disease characterized by the progressive accumulation of mucinous ascites and peritoneal implants. The optimal treatment for PMP includes the association of complete cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC). For patients with a large burdensome disease, the completeness of cytoreduction sometimes requires maximal effort surgery. The aim of this article is to provide proof of concept for two stage cytoreductive surgery (CRS) in this category of patients.Methods and materials: A two stage CRS and HIPEC with oxaliplatin was proposed for patients with bulky PMP including important involvement of the serosal surfaces of the bowel or colon who had an impaired nutritional status. The residual disease at the end of the first stage was less than 5 mm of thickness on several implants. Clinical, surgical and histopathological variables were analyzed.Results: All eight patients completed the two-stage strategy. Mortality was nil. One Clavien Dindo grade 3 event occurred in each stage. After a median follow up of 29.5 months, all patients were alive and free of recurrence. All of the patients had histopathological complete response on the specimens obtained from the residual sites during the second stage surgery.Conclusions: Two-stage surgical strategy is feasible for bulky PMP patients and it is associated with little high-grade morbidity and enhanced visceral sparing.
Collapse
Affiliation(s)
- Olivia Sgarbura
- Surgical Oncology Department, Montpellier Cancer Institute, University of Montpellier, Montpellier, France.,University of Medicine Montpellier 2 Rue de l'École de Médecine, Montpellier, France
| | - Mohammed Al Hosni
- Surgical Oncology Division, Department of Surgery, Sultan Qaboos University Hospital, Muscat, Oman
| | | | | | - Lakhdar Khellaf
- University of Medicine Montpellier 2 Rue de l'École de Médecine, Montpellier, France.,Pathology Department, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Marie-Hélène Pissas
- Surgical Oncology Department, Montpellier Cancer Institute, University of Montpellier, Montpellier, France.,University of Medicine Montpellier 2 Rue de l'École de Médecine, Montpellier, France
| | - Sébastien Carrère
- Surgical Oncology Department, Montpellier Cancer Institute, University of Montpellier, Montpellier, France.,University of Medicine Montpellier 2 Rue de l'École de Médecine, Montpellier, France
| | - Stephanie Nougaret
- University of Medicine Montpellier 2 Rue de l'École de Médecine, Montpellier, France.,Radiology Department, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Frédéric Bibeau
- Pathology Department, Caen University Hospital, University of Caen Normandy, Caen, France
| | - François Quénet
- Surgical Oncology Department, Montpellier Cancer Institute, University of Montpellier, Montpellier, France.,University of Medicine Montpellier 2 Rue de l'École de Médecine, Montpellier, France
| |
Collapse
|
38
|
Morgan R, Tun S, Eng OS. Hyperthermic Intraperitoneal Chemotherapy: An Overview #412. J Palliat Med 2021; 24:621-622. [PMID: 33797986 DOI: 10.1089/jpm.2020.0758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
39
|
Bhatt A, de Hingh I, Van Der Speeten K, Hubner M, Deraco M, Bakrin N, Villeneuve L, Kusamura S, Glehen O. HIPEC Methodology and Regimens: The Need for an Expert Consensus. Ann Surg Oncol 2021; 28:9098-9113. [PMID: 34142293 DOI: 10.1245/s10434-021-10193-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hyperthermic intraperitoneal chemotherapy (HIPEC) is performed with a wide variation in methodology, drugs, and other elements vital to the procedure. Adoption of a limited number of regimens could increase the collective experience of peritoneal oncologists, make comparison between studies more meaningful, and lead to a greater acceptance of results from randomized trials. This study aimed to determine the possibility of standardizing HIPEC methodology and regimens and to identify the best method of performing such a standardization. METHODS A critical review of preclinical and clinical studies evaluating the pharmacokinetic aspects of different HIPEC drugs and drug regimens, the impact of hyperthermia, and the efficacy of various HIPEC regimens as well as studies comparing different regimens was performed. RESULTS The preclinical and clinical data were limited, and studies comparing different regimens were scarce. Many of the regimens were neither supported by preclinical rationale or data nor validated by a dose-escalating formal phase 1 trial. All the regimens were based on pharmacokinetic data and did not take chemosensitivity of peritoneal metastases into account. Personalized medicine approaches such as patient-derived tumor organoids could offer a solution to this problem, although clinical validation is likely to be challenging. CONCLUSIONS Apart from randomized trials, more translational research and phases 1 and 2 studies are needed. While waiting for better preclinical and clinical evidence, the best way to minimize heterogeneity is by an expert consensus that aims to identify and define a limited number of regimens for each indication and primary site. The choice of regimen then can be tailored to the patient profile and its expected toxicity and the methodology according regional factors.
Collapse
Affiliation(s)
- Aditi Bhatt
- Department of Surgical Oncology, Zydus Hospital, Ahmedabad, India
| | - Ignace de Hingh
- Department of Surgical Oncology, Catharina Hospital, Eidhoven, The Netherlands
| | | | - Martin Hubner
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Marcello Deraco
- Department of Surgical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Naoual Bakrin
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
| | - Laurent Villeneuve
- Department of Clinical Research, Hospices Civils de Lyon, Centre Hospitalier Lyon-sud, Lyon, France
| | - Shigeki Kusamura
- Department of Surgical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France.
| |
Collapse
|
40
|
Bijelic L, Ramos I, Goeré D. The Landmark Series: Surgical Treatment of Colorectal Cancer Peritoneal Metastases. Ann Surg Oncol 2021; 28:4140-4150. [PMID: 33969466 DOI: 10.1245/s10434-021-10049-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Peritoneal metastases (PM) are a form of metastatic spread affecting approximately 5-15% of colon cancer patients. The attitude towards management of peritoneal metastases has evolved from therapeutic nihilism towards a more comprehensive and multidisciplinary approach, in large part due to the development of cytoreductive surgery (CRS), usually coupled with heated intraperitoneal chemotherapy (HIPEC), along with the constant improvement of systemic chemotherapy of colorectal cancer. Several landmark studies, including 5 randomized controlled trials have marked the development and refinement of surgical approaches to treating colorectal cancer peritoneal metastases. METHODS This review article focuses on these landmark studies and their influence in 4 key areas: the evidence supporting surgical resection of peritoneal metastases, the identification and standardization of important prognostic variables influencing patient selection, the role of surgery and intraperitoneal chemotherapy in prevention of colorectal PM and the role of intraperitoneal chemotherapy as an adjuvant to surgical resection. RESULTS These landmark studies indicate that surgical resection of colorectal PM should be considered as a therapeutic option in appropriately selected patients and when adequate surgical expertise is available. Standardized prognostic variables including the Peritoneal Cancer Index and the Completeness of Cytoreduction Score should be used for evaluating both indications and outcomes. CONCLUSIONS Current evidence does not support the use of second look surgery with oxaliplatin HIPEC or prophylactic oxaliplatin HIPEC in patients with high risk colon cancer nor the use of oxaliplatin HIPEC with CRS of colorectal PM.
Collapse
Affiliation(s)
- Lana Bijelic
- Peritoneal Malignancies Unit, Department of Surgery, Hospital Moises Broggi, Consorci Sanitari Integral, Barcelona, Spain.
| | - Isabel Ramos
- Peritoneal Malignancies Unit, Department of Surgery, Hospital Moises Broggi, Consorci Sanitari Integral, Barcelona, Spain
| | - Diane Goeré
- Department of Digestive and Oncological Surgery, Hôpital Saint-Louis - APHP, Université de Paris, Paris, France
| |
Collapse
|
41
|
Quénet F, Roca L, de Forges H, Gléhen O, Goéré D. Limitations of the PRODIGE 7 trial - Authors' reply. Lancet Oncol 2021; 22:e179-e180. [PMID: 33932377 DOI: 10.1016/s1470-2045(21)00192-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 01/06/2023]
Affiliation(s)
- François Quénet
- Department of Surgical Oncology, Institut du Cancer de Montpellier, University of Montpellier, Montpellier 34298, France.
| | - Lise Roca
- Biometrics Unit, Institut du Cancer de Montpellier, University of Montpellier, Montpellier 34298, France
| | - Hélène de Forges
- Department of Clinical Research and Innovation, Institut du Cancer de Montpellier, University of Montpellier, Montpellier 34298, France
| | - Olivier Gléhen
- Department of Digestive Surgery, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Diane Goéré
- Department of Digestive Surgery Hôpital Saint Louis, Paris, France
| |
Collapse
|
42
|
Lurvink RJ, Rauwerdink P, Rovers KP, Wassenaar ECE, Deenen MJ, Nederend J, Huysentruyt CJR, van 't Erve I, Fijneman RJA, van der Hoeven EJRJ, Seldenrijk CA, Constantinides A, Kranenburg O, Los M, Herbschleb KH, Thijs AMJ, Creemers GJM, Burger JWA, Wiezer MJ, Nienhuijs SW, Boerma D, de Hingh IHJT. First-line palliative systemic therapy alternated with electrostatic pressurised intraperitoneal aerosol chemotherapy (oxaliplatin) for isolated unresectable colorectal peritoneal metastases: protocol of a multicentre, single-arm, phase II study (CRC-PIPAC-II). BMJ Open 2021; 11:e044811. [PMID: 33785492 PMCID: PMC8011718 DOI: 10.1136/bmjopen-2020-044811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Despite its increasing use, first-line palliative systemic therapy alternated with electrostatic pressurised intraperitoneal aerosol chemotherapy with oxaliplatin (ePIPAC-OX), hereinafter referred to as first-line bidirectional therapy, has never been prospectively investigated in patients with colorectal peritoneal metastases (CPM). As a first step to address this evidence gap, the present study aims to assess the safety, feasibility, antitumour activity, patient-reported outcomes, costs and systemic pharmacokinetics of first-line bidirectional therapy in patients with isolated unresectable CPM. METHODS AND ANALYSIS In this single-arm, phase II study in two Dutch tertiary referral centres, 20 patients are enrolled. Key eligibility criteria are a good performance status, pathologically proven isolated unresectable CPM, no previous palliative systemic therapy for colorectal cancer, no (neo)adjuvant systemic therapy ≤6 months prior to enrolment and no previous pressurised intraperitoneal aerosol chemotherapy (PIPAC). Patients receive three cycles of bidirectional therapy. Each cycle consists of 6 weeks first-line palliative systemic therapy at the medical oncologists' decision (CAPOX-bevacizumab, FOLFOX-bevacizumab, FOLFIRI-bevacizumab or FOLFOXIRI-bevacizumab) followed by ePIPAC-OX (92 mg/m2) with an intraoperative bolus of intravenous leucovorin (20 mg/m2) and 5-fluorouracil (400 mg/m2). Study treatment ends after the third ePIPAC-OX. The primary outcome is the number of patients with-and procedures leading to-grade ≥3 adverse events (Common Terminology Criteria for Adverse Events V.5.0) up to 4 weeks after the last procedure. Key secondary outcomes include the number of bidirectional cycles in each patient, treatment-related characteristics, grade ≤2 adverse events, tumour response (histopathological, cytological, radiological, biochemical, macroscopic and ascites), patient-reported outcomes, systemic pharmacokinetics of oxaliplatin, costs, progression-free survival and overall survival. ETHICS AND DISSEMINATION This study is approved by the Dutch competent authority, a medical ethics committee and the institutional review boards of both study centres. Results will be submitted for publication in peer-reviewed medical journals and presented to patients and healthcare professionals. TRIAL REGISTRATION NUMBER NL8303.
Collapse
Affiliation(s)
- Robin J Lurvink
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Paulien Rauwerdink
- Department of Surgery, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Koen P Rovers
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Emma C E Wassenaar
- Department of Surgery, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, The Netherlands
| | - Joost Nederend
- Department of Radiology, Catharina Hospital, Eindhoven, The Netherlands
| | | | - Iris van 't Erve
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Remond J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | - Onno Kranenburg
- Department of Imaging and Cancer, UMC Utrecht, Utrecht, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Karin H Herbschleb
- Department of Medical Oncology, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Anna M J Thijs
- Department of Medical Oncology, Catharina Hospital, Eindhoven, The Netherlands
| | | | | | - Marinus J Wiezer
- Department of Surgery, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Simon W Nienhuijs
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Djamila Boerma
- Department of Surgery, Sint Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands
- School for Oncology and Developmental Biology, GROW, Maastricht, The Netherlands
| |
Collapse
|
43
|
Rovers KP, Lurvink RJ, Deenen MJ, de Hingh IHJT. RE: Phase I dose escalation study of oxaliplatin delivered via a laparoscopic approach using pressurized intraperitoneal aerosol chemotherapy (PIPAC) for advanced peritoneal metastases of gastrointestinal tract cancers: PIPAC-oxaliplatin for peritoneal metastases of gastrointestinal cancers. Eur J Cancer 2021; 147:182-184. [PMID: 33640212 DOI: 10.1016/j.ejca.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/12/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Koen P Rovers
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Robin J Lurvink
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Cancer Institute, PO Box 1350, 5602 ZA, Eindhoven, Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA, Eindhoven, Netherlands; GROW-School for Oncology and Developmental Biology, Maastricht University, PO Box 616, 6200 MD, Maastricht, Netherlands.
| |
Collapse
|
44
|
A letter of response to comments on 'A phase I dose-escalation study of oxaliplatin delivered via a laparoscopic approach using pressurised intraperitoneal aerosol chemotherapy for advanced peritoneal metastases of gastrointestinal tract cancers'. Eur J Cancer 2021; 147:185-186. [PMID: 33589368 DOI: 10.1016/j.ejca.2021.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 01/12/2021] [Indexed: 11/20/2022]
|
45
|
Rovers KP, Wassenaar ECE, Lurvink RJ, Creemers GJM, Burger JWA, Los M, Huysentruyt CJR, van Lijnschoten G, Nederend J, Lahaye MJ, Deenen MJ, Wiezer MJ, Nienhuijs SW, Boerma D, de Hingh IHJT. Pressurized Intraperitoneal Aerosol Chemotherapy (Oxaliplatin) for Unresectable Colorectal Peritoneal Metastases: A Multicenter, Single-Arm, Phase II Trial (CRC-PIPAC). Ann Surg Oncol 2021; 28:5311-5326. [PMID: 33544279 DOI: 10.1245/s10434-020-09558-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Despite its increasing use, pressurized intraperitoneal aerosol chemotherapy with oxaliplatin (PIPAC-OX) has never been prospectively investigated as a palliative monotherapy for colorectal peritoneal metastases in clinical trials. This trial aimed to assess the safety (primary aim) and antitumor activity (key secondary aim) of PIPAC-OX monotherapy in patients with unresectable colorectal peritoneal metastases. METHODS In this two-center, single-arm, phase II trial, patients with isolated unresectable colorectal peritoneal metastases in any line of palliative treatment underwent 6-weekly PIPAC-OX (92 mg/m2). Key outcomes were major treatment-related adverse events (primary outcome), minor treatment-related adverse events, hospital stay, tumor response (radiological, biochemical, pathological, ascites), progression-free survival, and overall survival. RESULTS Twenty enrolled patients underwent 59 (median 3, range 1-6) PIPAC-OX procedures. Major treatment-related adverse events occurred in 3 of 20 (15%) patients after 5 of 59 (8%) procedures (abdominal pain, intraperitoneal hemorrhage, iatrogenic pneumothorax, transient liver toxicity), including one possibly treatment-related death (sepsis of unknown origin). Minor treatment-related adverse events occurred in all patients after 57 of 59 (97%) procedures, the most common being abdominal pain (all patients after 88% of procedures) and nausea (65% of patients after 39% of procedures). Median hospital stay was 1 day (range 0-3). Response rates were 0% (radiological), 50% (biochemical), 56% (pathological), and 56% (ascites). Median progression-free and overall survival were 3.5 months (interquartile range [IQR] 2.5-5.7) and 8.0 months (IQR 6.3-12.6), respectively. CONCLUSIONS In patients with unresectable colorectal peritoneal metastases undergoing PIPAC-OX monotherapy, some major adverse events occurred and minor adverse events were common. The clinical relevance of observed biochemical, pathological, and ascites responses remains to be determined, especially since radiological response was absent.
Collapse
Affiliation(s)
- Koen P Rovers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Emma C E Wassenaar
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Robin J Lurvink
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Geert-Jan M Creemers
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Jacobus W A Burger
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, St. Antonius Hospital, Nieuwegein, The Netherlands
| | | | | | - Joost Nederend
- Department of Radiology, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Max J Lahaye
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Marinus J Wiezer
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Simon W Nienhuijs
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands. .,GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
46
|
Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy versus cytoreductive surgery alone for colorectal peritoneal metastases (PRODIGE 7): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2021; 22:256-266. [PMID: 33476595 DOI: 10.1016/s1470-2045(20)30599-4] [Citation(s) in RCA: 501] [Impact Index Per Article: 125.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The addition of hyperthermic intraperitoneal chemotherapy (HIPEC) to cytoreductive surgery has been associated with encouraging survival results in some patients with colorectal peritoneal metastases who were eligible for complete macroscopic resection. We aimed to assess the specific benefit of adding HIPEC to cytoreductive surgery compared with receiving cytoreductive surgery alone. METHODS We did a randomised, open-label, phase 3 trial at 17 cancer centres in France. Eligible patients were aged 18-70 years and had histologically proven colorectal cancer with peritoneal metastases, WHO performance status of 0 or 1, a Peritoneal Cancer Index of 25 or less, and were eligible to receive systemic chemotherapy for 6 months (ie, they had adequate organ function and life expectancy of at least 12 weeks). Patients in whom complete macroscopic resection or surgical resection with less than 1 mm residual tumour tissue was completed were randomly assigned (1:1) to cytoreductive surgery with or without oxaliplatin-based HIPEC. Randomisation was done centrally using minimisation, and stratified by centre, completeness of cytoreduction, number of previous systemic chemotherapy lines, and timing of protocol-mandated systemic chemotherapy. Oxaliplatin HIPEC was administered by the closed (360 mg/m2) or open (460 mg/m2) abdomen techniques, and systemic chemotherapy (400 mg/m2 fluorouracil and 20 mg/m2 folinic acid) was delivered intravenously 20 min before HIPEC. All individuals received systemic chemotherapy (of investigators' choosing) with or without targeted therapy before or after surgery, or both. The primary endpoint was overall survival, which was analysed in the intention-to-treat population. Safety was assessed in all patients who received surgery. This trial is registed with ClinicalTrials.gov, NCT00769405, and is now completed. FINDINGS Between Feb 11, 2008, and Jan 6, 2014, 265 patients were included and randomly assigned, 133 to the cytoreductive surgery plus HIPEC group and 132 to the cytoreductive surgery alone group. After median follow-up of 63·8 months (IQR 53·0-77·1), median overall survival was 41·7 months (95% CI 36·2-53·8) in the cytoreductive surgery plus HIPEC group and 41·2 months (35·1-49·7) in the cytoreductive surgery group (hazard ratio 1·00 [95·37% CI 0·63-1·58]; stratified log-rank p=0·99). At 30 days, two (2%) treatment-related deaths had occurred in each group.. Grade 3 or worse adverse events at 30 days were similar in frequency between groups (56 [42%] of 133 patients in the cytoreductive surgery plus HIPEC group vs 42 [32%] of 132 patients in the cytoreductive surgery group; p=0·083); however, at 60 days, grade 3 or worse adverse events were more common in the cytoreductive surgery plus HIPEC group (34 [26%] of 131 vs 20 [15%] of 130; p=0·035). INTERPRETATION Considering the absence of an overall survival benefit after adding HIPEC to cytoreductive surgery and more frequent postoperative late complications with this combination, our data suggest that cytoreductive surgery alone should be the cornerstone of therapeutic strategies with curative intent for colorectal peritoneal metastases. FUNDING Institut National du Cancer, Programme Hospitalier de Recherche Clinique du Cancer, Ligue Contre le Cancer.
Collapse
|
47
|
Dumont F, Passot C, Raoul JL, Kepenekian V, Lelièvre B, Boisdron-Celle M, Hiret S, Senellart H, Pein F, Blanc-Lapierre A, Raimbourg J, Thibaudeau E, Glehen O. A phase I dose-escalation study of oxaliplatin delivered via a laparoscopic approach using pressurised intraperitoneal aerosol chemotherapy for advanced peritoneal metastases of gastrointestinal tract cancers. Eur J Cancer 2020; 140:37-44. [PMID: 33039812 DOI: 10.1016/j.ejca.2020.09.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The objectives were to define the maximum tolerated dose (MTD), safety profile and pharmacokinetics (PKs) of intraperitoneal oxaliplatin delivered by pressurised intraperitoneal aerosol chemotherapy (PIPAC) in patients with advanced peritoneal carcinomatosis from gastrointestinal tract cancers. METHODS PIPAC was applied every 4-6 weeks, for 5 cycles, in a phase I dose-escalation study using a 3 + 3 design. The first dose level was 90 mg/m2 with planned increases of 50 mg/m2 per level. Platinum concentration was measured in plasma, tissues and intraperitoneal fluid samples. The trial was registered at ClinicalTrials.gov (NCT03294252). RESULTS Ten patients with 33 PIPAC sessions were included. No dose limiting toxicity (DLT) occurred at 90 mg/m2 and two at 140 mg/m2. The MTD was therefore set at 90 mg/m2. Overall treatment included a median number of three PIPAC sessions (range: 1-5) and secondary complete cytoreductive surgery for two patients. Overall safety showed 67 grade I-II and 11 grade III-IV toxicities, usually haematologic, digestive (nausea/vomiting, abdominal pain), and fatigue. Oxaliplatin concentrations were three- to four-fold higher in tissue in contact with aerosol than in muscle without contact. At 140 mg/m2, the plasma oxaliplatin concentration was high with Cmax and area under the curve (AUC)0-48h of 1035 μg/l and 9028 μg h/L, respectively. CONCLUSIONS The MTD of oxaliplatin during PIPAC is 90 mg/m2. PK data demonstrate a high tumour concentration and a significant systemic absorption.
Collapse
Affiliation(s)
- Frédéric Dumont
- Department of Surgical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France.
| | - Christophe Passot
- Oncopharmacology - Pharmacogenetics, Institut de Cancérologie de L'Ouest, Angers, France
| | - Jean-Luc Raoul
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Vahan Kepenekian
- Department of Surgical Oncology, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Bénédicte Lelièvre
- Laboratory of Pharmacology and Toxicology, Centre Hospitalier et Universitaire, Angers, France
| | | | - Sandrine Hiret
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Hélène Senellart
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Francois Pein
- Department of Clinical Research and Innovation, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Audrey Blanc-Lapierre
- Department of Biostatistics and Methodology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Judith Raimbourg
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Emilie Thibaudeau
- Department of Surgical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| |
Collapse
|
48
|
Narasimhan V, Flood M, Warrier S, Heriot A. Hyperthermic intraperitoneal chemotherapy for colorectal peritoneal metastases: still a necessity? ANZ J Surg 2020; 90:1541-1542. [PMID: 32924302 DOI: 10.1111/ans.15794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/19/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Vignesh Narasimhan
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Flood
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Satish Warrier
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alexander Heriot
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Lee SY, Fiorentini G, Szasz AM, Szigeti G, Szasz A, Minnaar CA. Quo Vadis Oncological Hyperthermia (2020)? Front Oncol 2020; 10:1690. [PMID: 33014841 PMCID: PMC7499808 DOI: 10.3389/fonc.2020.01690] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022] Open
Abstract
Heating as a medical intervention in cancer treatment is an ancient approach, but effective deep heating techniques are lacking in modern practice. The use of electromagnetic interactions has enabled the development of more reliable local-regional hyperthermia (LRHT) techniques whole-body hyperthermia (WBH) techniques. Contrary to the relatively simple physical-physiological concepts behind hyperthermia, its development was not steady, and it has gone through periods of failures and renewals with mixed views on the benefits of heating seen in the medical community over the decades. In this review we study in detail the various techniques currently available and describe challenges and trends of oncological hyperthermia from a new perspective. Our aim is to describe what we believe to be a new and effective approach to oncologic hyperthermia, and a change in the paradigm of dosing. Physiological limits restrict the application of WBH which has moved toward the mild temperature range, targeting immune support. LRHT does not have a temperature limit in the tumor (which can be burned out in extreme conditions) but a trend has started toward milder temperatures with immune-oriented goals, developing toward immune modulation, and especially toward tumor-specific immune reactions by which LRHT seeks to target the malignancy systemically. The emerging research of bystander and abscopal effects, in both laboratory investigations and clinical applications, has been intensified. Our present review summarizes the methods and results, and discusses the trends of hyperthermia in oncology.
Collapse
Affiliation(s)
- Sun-Young Lee
- Department of Radiation Oncology, Chonbuk National University Hospital, Jeonbuk, South Korea
| | | | - Attila Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Gyula Szigeti
- Innovation Center, Semmelweis University, Budapest, Hungary
| | - Andras Szasz
- Biotechnics Department, St. Istvan University, Godollo, Hungary
| | - Carrie Anne Minnaar
- Department of Radiation Oncology, Wits Donald Gordon Medical Center, Johannesburg, South Africa
| |
Collapse
|
50
|
Preparation and Evaluation of Intraperitoneal Long-Acting Oxaliplatin-Loaded Multi-Vesicular Liposomal Depot for Colorectal Cancer Treatment. Pharmaceutics 2020; 12:pharmaceutics12080736. [PMID: 32764318 PMCID: PMC7466130 DOI: 10.3390/pharmaceutics12080736] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer with peritoneal metastasis has a poor prognosis because of inadequate responses to systemic chemotherapy. Cytoreductive surgery followed by intraperitoneal (IP) chemotherapy using oxaliplatin has attracted attention; however, the short half-life of oxaliplatin and its rapid clearance from the peritoneal cavity limit its clinical application. Here, a multivesicular liposomal (MVL) depot of oxaliplatin was prepared for IP administration, with an expected prolonged effect. After optimization, a combination of phospholipids, cholesterol, and triolein was used based on its ability to produce MVL depots of monomodal size distribution (1–20 µm; span 1.99) with high entrapment efficiency (EE) (92.16% ± 2.17%). An initial burst release followed by a long lag phase of drug release was observed for the MVL depots system in vitro. An in vivo pharmacokinetic study mimicking the early postoperative IP chemotherapy regimen in rats showed significantly improved bioavailability, and the mean residence time of oxaliplatin after IP administration revealed that slow and continuous erosion of the MVL particles yielded a sustained drug release. Thus, oxaliplatin-loaded MVL depots presented in this study have potential for use in the treatment of colorectal cancer.
Collapse
|