1
|
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
|
2
|
Cortés-Guiral D, Kranenburg O, Sgarbura O, Van Der Speeten K, Taibi A, Hübner M, Yacoov AB. PIPAC Pharmacologic and Clinical Data. J Surg Oncol 2024; 130:1337-1348. [PMID: 39315493 DOI: 10.1002/jso.27900] [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: 08/20/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024]
Abstract
Pressurized intraperitoneal aerosol chemotherapy (PIPAC) emerged as an innovative intraperitoneal chemotherapy delivery system to overcome the issue of limited efficacy of systemic therapies to induce response in peritoneal malignancies. Promising results for patients with mesothelioma peritonei and peritoneal metastasis from gastric, ovarian, colorectal, pancreatic, and hepatobiliary tumors origin are changing the landscape for patients otherwise just facing palliative treatment. Ongoing trials will shed more light on the actual benefits of PIPAC.
Collapse
Affiliation(s)
- Delia Cortés-Guiral
- IVOQA (Viamed Advanced Surgical Oncology Institute), Hospital Viamed Santa Elena, Madrid, Spain
| | - Onno Kranenburg
- Lab Translational Oncology Cancer, Department of Surgical Oncology, Regenerative Medicine and Stem Cells, Utrecht Platform for Organoid Technology (UPORT), UMCU, Utrecht, The Netherlands
- Laboratory of Translational Oncology, Division of Imaging and Cancer, UMCU, Utrecht, The Netherlands
| | - Olivia Sgarbura
- Department of Surgical Oncology, Cancer Institute Montpellier, Montpellier, France
| | - Kurt Van Der Speeten
- Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium
- Faculty of Medicine and Life Sciences, BIOMED Research Institute, University Hasselt, Hasselt, Belgium
| | - Albdelkader Taibi
- Digestive Surgery Department, Dupuytren Limoges University Hospital, Limoges, France. CNRS, XLIM, UMR 7252, University Limoges, Limoges, France
| | - Martin Hübner
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Almog Ben Yacoov
- Department of General Surgery C and Surgical Oncology, Sheba Medical Center, Ramat Gan, Israel, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
3
|
Van der Speeten K, Kusamura S, Villeneuve L, Piso P, Verwaal VJ, González-Moreno S, Glehen O. The 2022 PSOGI International Consensus on HIPEC Regimens for Peritoneal Malignancies: HIPEC Technologies. Ann Surg Oncol 2024; 31:7090-7110. [PMID: 39037523 DOI: 10.1245/s10434-024-15513-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/09/2024] [Indexed: 07/23/2024]
Abstract
This manuscript reports the results of an international consensus on technologies of hyperthermic intraperitoneal perioperative chemotherapy (HIPEC) performed with the following goals: To provide recommendations for the technological parameters to perform HIPEC. To identify the role of heat and its application forms in treating peritoneal metastases. To provide recommendations regarding the correct dosimetry of intraperitoneal chemotherapy drugs and their carrier solutions. To identify for each intraperitoneal chemotherapy regimen the best dosimetry and fractionation. To identify areas of future research pertaining to HIPEC technology and regimens. This consensus was performed by the Delphi technique and comprised two rounds of voting. In total, 96 of 102 eligible panelists replied to both Delphi rounds (94.1%) with a consensus of 39/51 questions on HIPEC technical aspects. Among the recommendations that met with the strongest consensus were those concerning the dose of HIPEC drug established in mg/m2, a target temperature of at least 42°C, and the use of at least three temperature probes to pursue hyperthermia. Ninety minutes as the ideal HIPEC duration seemed to make consensus. These results should be considered when designing new clinical trials in patients with peritoneal surface malignancies.
Collapse
Affiliation(s)
- Kurt Van der Speeten
- Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium.
- Faculty of Life Sciences, BIOMED Research Institute, University Hasselt, Hasselt, Belgium.
| | - Shigeki Kusamura
- Department of Surgical Oncology, PSM unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laurent Villeneuve
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
| | - Pompiliu Piso
- Department of General and Visceral Surgery, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Vic J Verwaal
- Peritoneal Surface Malignancy and HIPEC Institute for Regional Sundhedforskning, Syddansk University, Odense, Sweden
| | | | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
| |
Collapse
|
4
|
Shao L, Wang N, Yan Y, Tan Y, Wu Q, Lei L, Wang M, Liu L. Quercetin of huoxuehuayu tongluo decoction and azithromycin combination therapy effectively improves rat tubal factor infertility by inhibiting inflammation. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:685-694. [PMID: 38645489 PMCID: PMC11024413 DOI: 10.22038/ijbms.2024.72049.15662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/13/2023] [Indexed: 04/23/2024]
Abstract
Objectives Tubal factor infertility (TFI) is common female infertility responsible for a large portion of female factor infertility. This study reveals the effect of the quercetin of Huoxuehuayu Tongluo Decoction with azithromycin on the pregnancy rate and inflammation of TFI female rats. Materials and Methods Female Sprague Dawley rats were constructed into the TFI model and treated with quercetin, Huoxuehuayu Tongluo Decoction, and combination therapy (quercetin and azithromycin). Pregnancy rate and litter size were measured. Network pharmacology was applied to analyze the interaction between Huoxuehuayu Tongluo Decoction and TFI. The combination of quercetin and IL-6 was analyzed by molecular docking. HE staining and electron microscopy were used to observe the histopathology and ultrastructure of fallopian tube tissues. The TNF-α, IL-1β, IL-6, IL-8, and MPO levels were detected by ELISA. The activation of JAK/STAT, MAPK, and NF-κB p65 pathways was detected by western blot or immunohistochemistry. Results Quercetin was the main active component of Huoxuehuayu Tongluo Decoction, and could bind to IL-6 in TFI. Target genes were enriched in the IL-17 signaling pathway, JAK-STAT signaling pathway, inflammatory disease, etc. Under the quercetin and azithromycin combination therapy, both rat pregnancy rates and litter sizes increased significantly. quercetin and azithromycin alleviated the symptoms of hydrosalpinx and inflammatory damage in fallopian tube tissues. The phosphorylation of JAK/STAT and MAPK pathways and NF-κB p65 translocation to the nucleus were significantly inhibited by the quercetin and azithromycin therapy. Conclusion Quercetin and azithromycin combination therapy inhibited inflammation and phosphorylation of JAK/STAT and MAPK pathways to improve TFI inflammation and pregnancy function.
Collapse
Affiliation(s)
- Liang Shao
- Department of Emergency, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| | - Nansu Wang
- Department of TCM Gynaecology, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| | - Yan Yan
- Department of TCM Gynaecology, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| | - Yali Tan
- Department of TCM Gynaecology, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| | - Qin Wu
- Department of TCM Gynaecology, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| | - Lei Lei
- Combine traditional Chinese and Western Medicine Institute, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Mingming Wang
- Clinical Laboratory, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| | - Ling Liu
- Department of TCM Gynaecology, the First Affiliated Hospital of Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
| |
Collapse
|
5
|
Hübner M, van Der Speeten K, Govaerts K, de Hingh I, Villeneuve L, Kusamura S, Glehen O. 2022 Peritoneal Surface Oncology Group International Consensus on HIPEC Regimens for Peritoneal Malignancies: Colorectal Cancer. Ann Surg Oncol 2024; 31:567-576. [PMID: 37940803 PMCID: PMC10695877 DOI: 10.1245/s10434-023-14368-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/13/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Selected patients with peritoneal metastases of colorectal cancer (PM-CRC) can benefit from potentially curative cytoreductive surgery (CRS) ± hyperthermic intraperitoneal chemotherapy (HIPEC), with a median overall survival (OS) of more than 40 months. OBJECTIVE The aims of this evidence-based consensus were to define the indications for HIPEC, to select the preferred HIPEC regimens, and to define research priorities regarding the use of HIPEC for PM-CRC. METHODS The consensus steering committee elaborated and formulated pertinent clinical questions according to the PICO (patient, intervention, comparator, outcome) method and assessed the evidence according to the Grading of Recommendation, Assessment, Development, and Evaluation (GRADE) framework. Standardized evidence tables were presented to an international expert panel to reach a consensus (4-point, weak and strong positive/negative) on HIPEC regimens and research priorities through a two-round Delphi process. The consensus was defined as ≥ 50% agreement for the 4-point consensus grading or ≥ 70% for either of the two combinations. RESULTS Evidence was weak or very weak for 9/10 clinical questions. In total, 70/90 eligible panelists replied to both Delphi rounds (78%), with a consensus for 10/10 questions on HIPEC regimens. There was strong negative consensus concerning the short duration, high-dose oxaliplatin (OX) protocol (55.7%), and a weak positive vote (53.8-64.3%) in favor of mitomycin-C (MMC)-based HIPEC (preferred choice: Dutch protocol: 35 mg/m2, 90 min, three fractions), both for primary cytoreduction and recurrence. Determining the role of HIPEC after CRS was considered the most important research question, regarded as essential by 85.7% of the panelists. Furthermore, over 90% of experts suggest performing HIPEC after primary and secondary CRS for recurrence > 1 year after the index surgery. CONCLUSIONS Based on the available evidence, despite the negative results of PRODIGE 7, HIPEC could be conditionally recommended to patients with PM-CRC after CRS. While more preclinical and clinical data are eagerly awaited to harmonize the procedure further, the MMC-based Dutch protocol remains the preferred regimen after primary and secondary CRS.
Collapse
Affiliation(s)
- Martin Hübner
- Department of Visceral Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Kurt van Der Speeten
- Department of Abdominal and Oncological Surgery, Ziekenhuis Oost Limburg (ZOL), Genk, Belgium
| | - Kim Govaerts
- Department of Abdominal and Oncological Surgery, Ziekenhuis Oost Limburg (ZOL), Genk, Belgium
| | - Ignace de Hingh
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Laurent Villeneuve
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | | | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
- CICLY: Center for Innovation in Cancer in Lyon, University Lyon 1, Lyon, France
| |
Collapse
|
6
|
Lv Q, Wang Y, Xiong Z, Xue Y, Li J, Chen M, Zhou K, Xu H, Zhang X, Liu J, Ren J, Liu B. Microvascularized tumor assembloids model for drug delivery evaluation in colorectal cancer-derived peritoneal metastasis. Acta Biomater 2023; 168:346-360. [PMID: 37393969 DOI: 10.1016/j.actbio.2023.06.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/27/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
Peritoneal metastasis (PM) is a fatal state of colorectal cancer, and only a few patients may benefit from systemic chemotherapy. Although hyperthermic intraperitoneal chemotherapy (HIPEC) brings hope for affected patients, the drug development and preclinical evaluation of HIPEC are seriously lagging behind, mainly due to the lack of an ideal in vitro PM model that makes drug development over-reliant on expensive and inefficient animal experiments. This study developed an in vitro colorectal cancer PM model [microvascularized tumor assembloids (vTA)] based on an assembly strategy of endothelialized microvessels and tumor spheroids. Our data showed that the in vitro perfusion cultured vTA could maintain a similar gene expression pattern to their parental xenografts. Also, the drug penetration pattern of the in vitro HIPEC in vTA could mimic the drug delivery behavior in tumor nodules during in vivo HIPEC. More importantly, we further confirmed the feasibility of constructing a tumor burden-controlled PM animal model using vTA. In conclusion, we propose a simple and effective strategy to construct physiologically simulated PM models in vitro, thus providing a basis for PM-related drug development and preclinical evaluation of locoregional therapies. STATEMENT OF SIGNIFICANCE: This study developed an in vitro colorectal cancer peritoneal metastasis (PM) model based on microvascularized tumor assembloids (vTA) for drug evaluation. With perfusion culture, vTA could maintain a similar gene expression pattern and tumor heterogeneity to their parental xenografts. And the drug penetration pattern in vTA was similar to the drug delivery behavior in tumor nodules under in vivo treatment. Moreover, vTA was more conducive to construct PM animal models with controllable tumor burden. In conclusion, the construction of vTA could provide a new strategy for the PM-related drug development and preclinical evaluation of locoregional therapies.
Collapse
Affiliation(s)
- Qijun Lv
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Ultrasound Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China; Department of Gastrointestinal Surgery, the Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yizhen Wang
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Zhiyong Xiong
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Yifan Xue
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Jiajun Li
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Moyang Chen
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Kaijian Zhou
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Hetao Xu
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Xiaoge Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Jie Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China.
| | - Jie Ren
- Department of Ultrasound Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China.
| | - Bo Liu
- Department of General Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
7
|
Helderman RFCPA, Bokan B, van Bochove GGW, Rodermond HM, Thijssen E, Marchal W, Torang A, Löke DR, Franken NAP, Kok HP, Tanis PJ, Crezee J, Oei AL. Elevated temperatures and longer durations improve the efficacy of oxaliplatin- and mitomycin C-based hyperthermic intraperitoneal chemotherapy in a confirmed rat model for peritoneal metastasis of colorectal cancer origin. Front Oncol 2023; 13:1122755. [PMID: 37007077 PMCID: PMC10064448 DOI: 10.3389/fonc.2023.1122755] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/21/2023] [Indexed: 03/19/2023] Open
Abstract
IntroductionIn patients with limited peritoneal metastasis (PM) originating from colorectal cancer, cytoreductive surgery (CRS) followed by hyperthermic intraperitoneal chemotherapy (HIPEC) is a potentially curative treatment option. This combined treatment modality using HIPEC with mitomycin C (MMC) for 90 minutes proved to be superior to systemic chemotherapy alone, but no benefit of adding HIPEC to CRS alone was shown using oxaliplatin-based HIPEC during 30 minutes. We investigated the impact of treatment temperature and duration as relevant HIPEC parameters for these two chemotherapeutic agents in representative preclinical models. The temperature- and duration- dependent efficacy for both oxaliplatin and MMC was evaluated in an in vitro setting and in a representative animal model.MethodsIn 130 WAG/Rij rats, PM were established through i.p. injections of rat CC-531 colon carcinoma cells with a signature similar to the dominant treatment-resistant CMS4 type human colorectal PM. Tumor growth was monitored twice per week using ultrasound, and HIPEC was applied when most tumors were 4-6 mm. A semi-open four-inflow HIPEC setup was used to circulate oxaliplatin or MMC through the peritoneum for 30, 60 or 90 minutes with inflow temperatures of 38°C or 42°C to achieve temperatures in the peritoneum of 37°C or 41°C. Tumors, healthy tissue and blood were collected directly or 48 hours after treatment to assess the platinum uptake, level of apoptosis and proliferation and to determine the healthy tissue toxicity.ResultsIn vitro results show a temperature- and duration- dependent efficacy for both oxaliplatin and MMC in both CC-531 cells and organoids. Temperature distribution throughout the peritoneum of the rats was stable with normothermic and hyperthermic average temperatures in the peritoneum ranging from 36.95-37.63°C and 40.51-41.37°C, respectively. Treatments resulted in minimal body weight decrease (<10%) and only 7/130 rats did not reach the endpoint of 48 hours after treatment.ConclusionsBoth elevated temperatures and longer treatment duration resulted in a higher platinum uptake, significantly increased apoptosis and lower proliferation in PM tumor lesions, without enhanced normal tissue toxicity. Our results demonstrated that oxaliplatin- and MMC-based HIPEC procedures are both temperature- and duration-dependent in an in vivo tumor model.
Collapse
Affiliation(s)
- Roxan F. C. P. A. Helderman
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Bella Bokan
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Gregor G. W. van Bochove
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Hans M. Rodermond
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Elsy Thijssen
- Institute for Materials Research, Analytical and Circular Chemistry, Hasselt University, Diepenbeek, Belgium
| | - Wouter Marchal
- Institute for Materials Research, Analytical and Circular Chemistry, Hasselt University, Diepenbeek, Belgium
| | - Arezo Torang
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Oncode Institute, Amsterdam, Netherlands
| | - Daan R. Löke
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Nicolaas A. P. Franken
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - H. Petra Kok
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Pieter J. Tanis
- Department of Surgery, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Arlene L. Oei
- Department of Radiation Oncology, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
- *Correspondence: Arlene L. Oei,
| |
Collapse
|
8
|
Witmer HDD, Dhiman A, Turaga KK. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for colorectal peritoneal metastases: Where do we stand? Cancer 2023; 129:495-502. [PMID: 36527271 PMCID: PMC10107206 DOI: 10.1002/cncr.34567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 12/23/2022]
Abstract
PLAIN LANGUAGE SUMMARY Patients with colorectal cancer that has spread to the lining of the abdomen (peritoneum) benefit from surgery to remove all the cancer. The addition of certain types of intra-abdominal chemotherapy during surgery improves survival for select patients.
Collapse
Affiliation(s)
- Hunter D D Witmer
- Department of Surgery, University of Chicago Medicine, Chicago, Illinois, USA
| | - Ankit Dhiman
- Department of Surgery, University of Chicago Medicine, Chicago, Illinois, USA
| | - Kiran K Turaga
- Department of Surgery, University of Chicago Medicine, Chicago, Illinois, USA
| |
Collapse
|
9
|
Liesenfeld LF, Quiring E, Al-Saeedi M, Nusshag C, Büchler MW, Schneider M. Extensive Peritonectomy is an Independent Risk Factor for Cisplatin HIPEC-Induced Acute Kidney Injury. Ann Surg Oncol 2022; 30:2646-2656. [PMID: 36496489 PMCID: PMC10085927 DOI: 10.1245/s10434-022-12661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/25/2022] [Indexed: 12/14/2022]
Abstract
Abstract
Background
Cisplatin (CDDP)-containing hyperthermic intraperitoneal chemotherapy (HIPEC) is frequently applied in selected patients with peritoneal malignancies derived from ovarian cancer, gastric cancer, and primary peritoneal mesothelioma. HIPEC with CDDP increases perioperative morbidity, in particular by inducing acute kidney injury (AKI). Factors contributing to occurrence of AKI after intraperitoneal perfusion with CDDP have not been sufficiently evaluated.
Patients and Methods
Data from 63 patients treated with a CDDP-containing HIPEC regimen were retrospectively analyzed concerning demographics, underlying disease, surgery, and HIPEC details to evaluate risk factors of AKI. A preclinical rat perfusion model was applied to assess the influence of temperature, concentration, perfusate volume, perfusion flow rate, and extent of peritonectomy on drug absorption upon intraperitoneal CDDP perfusion.
Results
AKI occurred in 66.1% of patients undergoing CDDP-containing HIPEC, with total intraoperative fluid influx being a negative and the extent of parietal peritonectomy being a positive independent predictor of postoperative AKI. In a preclinical model, bilateral anterior parietal peritonectomy significantly increased systemic CDDP absorption by 1.6 to 2-fold. CDDP plasma levels in animals were significantly higher after both perfusion with increased CDDP perfusate concentrations and bilateral anterior parietal peritonectomy.
Conclusion
CDDP-containing HIPEC is associated with relevant morbidity owing to its systemic toxicity. Extent of parietal peritonectomy is an independent predictor of AKI. CDDP dose reduction should be considered in case of extensive parietal peritonectomy. Cytostatic drug concentrations in HIPEC perfusate should be paid more attention to than total dose per body surface area. Further clinical studies are needed to confirm the presented preclinical findings.
Collapse
Affiliation(s)
- Lukas F Liesenfeld
- Department of General, Visceral and Transplantation Surgery, Medical Faculty, University Hospital Heidelberg, Heidelberg, Germany
| | - Eva Quiring
- Department of General, Visceral and Transplantation Surgery, Medical Faculty, University Hospital Heidelberg, Heidelberg, Germany
| | - Mohammed Al-Saeedi
- Department of General, Visceral and Transplantation Surgery, Medical Faculty, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, Medical Faculty, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, Medical Faculty, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, Medical Faculty, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
10
|
Kepenekian V, Bhatt A, Péron J, Alyami M, Benzerdjeb N, Bakrin N, Falandry C, Passot G, Rousset P, Glehen O. Advances in the management of peritoneal malignancies. Nat Rev Clin Oncol 2022; 19:698-718. [PMID: 36071285 DOI: 10.1038/s41571-022-00675-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2022] [Indexed: 11/09/2022]
Abstract
Peritoneal surface malignancies (PSMs) are usually associated with a poor prognosis. Nonetheless, in line with advances in the management of most abdominopelvic metastatic diseases, considerable progress has been made over the past decade. An improved understanding of disease biology has led to the more accurate prediction of neoplasia aggressiveness and the treatment response and has been reflected in the proposal of new classification systems. Achieving complete cytoreductive surgery remains the cornerstone of curative-intent treatment of PSMs. Alongside centralization in expert centres, enabling the delivery of multimodal and multidisciplinary strategies, preoperative management is a crucial step in order to select patients who are most likely to benefit from surgery. Depending on the specific PSM, the role of intraperitoneal chemotherapy and of perioperative systemic chemotherapy, in particular, in the neoadjuvant setting, is established in certain scenarios but questioned in several others, although more prospective data are required. In this Review, we describe advances in all aspects of the management of PSMs including disease biology, assessment and improvement of disease resectability, perioperative management, systemic therapy and pre-emptive management, and we speculate on future research directions.
Collapse
Affiliation(s)
- Vahan Kepenekian
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France
| | - Aditi Bhatt
- Department of Surgical Oncology, Zydus hospital, Ahmedabad, Gujarat, India
| | - Julien Péron
- Medical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique-Santé, UCBL1, Lyon, France
| | - Mohammad Alyami
- Department of General Surgery and Surgical Oncology, Oncology Center, King Khalid Hospital, Najran, Saudi Arabia
| | - Nazim Benzerdjeb
- CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France.,Department of Pathology, Institut de Pathologie Multisite, Hospices Civils de Lyon, UCBL1, Lyon, France
| | - Naoual Bakrin
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France
| | - Claire Falandry
- Department of Onco-Geriatry, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Guillaume Passot
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France
| | - Pascal Rousset
- CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France.,Department of Radiology, Hôpital Lyon Sud, Hospices Civils de Lyon, UCBL1, Lyon, France
| | - Olivier Glehen
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France. .,CICLY - EA3738, Université Claude Bernard Lyon I (UCBL1), Lyon, France.
| |
Collapse
|
11
|
Batista TP, Carneiro VCG, Tancredi R, Badiglian Filho L, Rangel RLC, Lopes A, Sarmento BJQ, Leão CS. A phase 2 trial of short-course Hyperthermic IntraPeritoneal Chemotherapy (HIPEC) at interval cytoreductive surgery (iCRS) for advanced ovarian cancer. Rev Col Bras Cir 2022; 49:e20223135. [PMID: 35239855 PMCID: PMC10578806 DOI: 10.1590/0100-6991e-20223135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/06/2021] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE to report the final analysis of a phase 2 trial assessing the efficacy and safety of short-course hyperthermic intraperitoneal chemotherapy (HIPEC) for patients with advanced epithelial ovarian cancer (EOC). METHODS this was an open-label, multicenter, single-arm trial of HIPEC in patients with advanced EOC who underwent interval cytoreductive surgery (iCRS) after neoadjuvant chemotherapy (NACT). HIPEC was performed as a concentration-based regimen of platinum-based chemotherapy for 30 minutes. Primary endpoint was the rate of disease progression occurring at nine months following iCRS plus HIPEC (PD9). Secondary endpoints were postoperative complications, time to start adjuvant chemotherapy, length of hospital and ICU stay, quality of life (QoL) over treatment, and ultimately 2-year progression-free survival (PFS) and overall survival (OS). Analysis was by intention-to-treat with final database lock for survival outcomes on February 23, 2021. RESULTS fifteen patients with stage III EOC were enrolled between February 2015 and July 2019, in four centers. The intention to treat PD9 was 6.7%. With a median follow-up of 33 months (IQR, 24.3-46.5), the median PFS was 18.1 months and corresponding 2-year rates of PFS and OS was 33.3% and 93.3%, respectively. Three patients (20%) experienced graded III complications. Median length of hospital and ICU stay was 5 (IQR, 4-6.5) and 1 (IQR, 1-1) days, respectively. Time to restart systemic chemotherapy was 39 (IQR, 35-49.3) days and no significant difference over time in QoL was observed. CONCLUSIONS we demonstrate preliminary efficacy and safety of short-course HIPEC in patient with advanced EOC.
Collapse
Affiliation(s)
- Thales Paulo Batista
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Department of Surgery/Oncology - Recife - PE - Brasil
- - UFPE - Universidade Federal de Pernambuco, Center of Medical Science - Recife - PE - Brasil
| | - Vandré Cabral Gomes Carneiro
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Department of Surgery/Oncology - Recife - PE - Brasil
- - HCP - Hospital de Câncer de Pernambuco, Department of Gynecology - Recife - PE - Brasil
| | - Rodrigo Tancredi
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Department of Clinical Oncology - Recife - PE - Brasil
- - HCP - Hospital de Câncer de Pernambuco, Department of Clinical Oncology - Recife - PE - Brasil
| | | | | | - André Lopes
- - IBCC - Instituto Brasileiro de Controle do Câncer, Department of Gynecology - São Paulo - SP - Brasil
| | - Bruno José Queiroz Sarmento
- - IHBDF - Instituto Hospital de Base do Distrito Federal, Serviço de Oncologia Cirúrgica - Brasília - DF - Brasil
| | - Cristiano Souza Leão
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Departamento de Cirurgia - Recife - PE - Brasil
| |
Collapse
|
12
|
Löke DR, Helderman RFCPA, Franken NAP, Oei AL, Tanis PJ, Crezee J, Kok HP. Simulating drug penetration during hyperthermic intraperitoneal chemotherapy. Drug Deliv 2021; 28:145-161. [PMID: 33427507 PMCID: PMC7808385 DOI: 10.1080/10717544.2020.1862364] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Hyperthermic intraperitoneal chemotherapy (HIPEC) is administered to treat residual microscopic disease after debulking cytoreductive surgery. During HIPEC, a limited number of catheters are used to administer and drain fluid containing chemotherapy (41–43 °C), yielding heterogeneities in the peritoneum. Large heterogeneities may lead to undertreated areas, increasing the risk of recurrences. Aiming at intra-abdominal homogeneity is therefore essential to fully exploit the potential of HIPEC. More insight is needed into the extent of the heterogeneities during treatments and assess their effects on the efficacy of HIPEC. To that end we developed a computational model containing embedded tumor nodules in an environment mimicking peritoneal conditions. Tumor- and treatment-specific parameters affecting drug delivery like tumor size, tumor shape, velocity, temperature and dose were assessed using three-dimensional computational fluid dynamics (CFD) to demonstrate their effect on the drug distribution and accumulation in nodules. Clonogenic assays performed on RKO colorectal cell lines yielded the temperature-dependent IC50 values of cisplatin (19.5–6.8 micromolar for 37–43 °C), used to compare drug distributions in our computational models. Our models underlined that large nodules are more difficult to treat and that temperature and velocity are the most important factors to control the drug delivery. Moderate flow velocities, between 0.01 and 1 m/s, are optimal for the delivery of cisplatin. Furthermore, higher temperatures and higher doses increased the effective penetration depth with 69% and 54%, respectively. We plan to extend the software developed for this study toward patient-specific treatment planning software, capable of mapping and assist in reducing heterogeneous flow patterns.
Collapse
Affiliation(s)
- Daan R Löke
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Roxan F C P A Helderman
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicolaas A P Franken
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Arlene L Oei
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter J Tanis
- Department for Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Vos LMC, Aronson SL, van Driel WJ, Huitema ADR, Schagen van Leeuwen JH, Lok CAR, Sonke GS. Translational and pharmacological principles of hyperthermic intraperitoneal chemotherapy for ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2021; 78:86-102. [PMID: 34565676 DOI: 10.1016/j.bpobgyn.2021.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022]
Abstract
The long-term survival of advanced-stage ovarian cancer patients remains poor, despite extensive cytoreductive surgery, chemotherapy, and the recent addition of poly (ADP-ribose) polymerase inhibitors (PARPi). Hyperthermic intraperitoneal chemotherapy (HIPEC) has shown survival benefit by specifically targeting peritoneal metastases, the primary site of disease recurrence. Different aspects of how HIPEC exerts its effect remain poorly understood. Improved understanding of the effects of hyperthermia on ovarian cancer cells, the synergy of hyperthermia with intraperitoneal chemotherapy, and the pharmacological and pharmacokinetic properties of intraperitoneally administered cisplatin may help identify ways to optimize the efficacy of HIPEC. This review provides an overview of these translational and pharmacological principles of HIPEC and aims to expose knowledge gaps that may direct further research to optimize the HIPEC procedure and ultimately improve survival for women with advanced ovarian cancer.
Collapse
Affiliation(s)
- Laura M C Vos
- Dept. of Gynecologic Oncology, Center for Gynecologic Oncology, Amsterdam, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - S Lot Aronson
- Dept. of Gynecologic Oncology, Center for Gynecologic Oncology, Amsterdam, Netherlands Cancer Institute, Amsterdam, the Netherlands; Dept. of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willemien J van Driel
- Dept. of Gynecologic Oncology, Center for Gynecologic Oncology, Amsterdam, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alwin D R Huitema
- Dept. of Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Dept. of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Dept. of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Christine A R Lok
- Dept. of Gynecologic Oncology, Center for Gynecologic Oncology, Amsterdam, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Gabe S Sonke
- Dept. of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Preclinical In Vivo-Models to Investigate HIPEC; Current Methodologies and Challenges. Cancers (Basel) 2021; 13:cancers13143430. [PMID: 34298644 PMCID: PMC8303745 DOI: 10.3390/cancers13143430] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Efficacy of cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) depends on patient selection, tumor type, delivery technique, and treatment parameters such as temperature, carrier solution, type of drug, dosage, volume, and treatment duration. Preclinical research offers a powerful tool to investigate the impact of these parameters and to assists in designing potentially more effective treatment protocols and clinical trials. This study aims to review the objectives, methods, and clinical relevance of in vivo preclinical HIPEC studies found in the literature. In total, 60 articles were included in this study. The selected articles were screened on the HIPEC parameters. Recommendations are provided and possible pitfalls are discussed on the choice of type of animal and tumor model per stratified parameters and study goal. The guidelines presented in this paper can improve the clinical relevance and impact of future in vivo HIPEC experiments. Abstract Hyperthermic intraperitoneal chemotherapy (HIPEC) is a treatment modality for patients with peritoneal metastasis (PM) of various origins which aims for cure in combination with cytoreductive surgery (CRS). Efficacy of CRS-HIPEC depends on patient selection, tumor type, delivery technique, and treatment parameters such as temperature, carrier solution, type of drug, dosage, volume, and treatment duration. Preclinical research offers a powerful tool to investigate the impact of these parameters and to assist in designing potentially more effective treatment protocols and clinical trials. The different methodologies for peritoneal disease and HIPEC are variable. This study aims to review the objectives, methods, and clinical relevance of in vivo preclinical HIPEC studies found in the literature. In this review, recommendations are provided and possible pitfalls are discussed on the choice of type of animal and tumor model per stratified parameters and study goal. The guidelines presented in this paper can improve the clinical relevance and impact of future in vivo HIPEC experiments.
Collapse
|
15
|
Löke DR, Helderman RFCPA, Rodermond HM, Tanis PJ, Streekstra GJ, Franken NAP, Oei AL, Crezee J, Kok HP. Demonstration of treatment planning software for hyperthermic intraperitoneal chemotherapy in a rat model. Int J Hyperthermia 2021; 38:38-54. [PMID: 33487083 DOI: 10.1080/02656736.2020.1852324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Hyperthermic intraperitoneal chemotherapy (HIPEC) is administered to treat residual microscopic disease after cytoreductive surgery (CRS). During HIPEC, fluid (41-43 °C) is administered and drained through a limited number of catheters, risking thermal and drug heterogeneities within the abdominal cavity that might reduce effectiveness. Treatment planning software provides a unique tool for optimizing treatment delivery. This study aimed to investigate the influence of treatment-specific parameters on the thermal and drug homogeneity in the peritoneal cavity in a computed tomography based rat model. METHOD We developed computational fluid dynamics (CFD) software simulating the dynamic flow, temperature and drug distribution during oxaliplatin based HIPEC. The influence of location and number of catheters, flow alternations and flow rates on peritoneal temperature and drug distribution were determined. The software was validated using data from experimental rat HIPEC studies. RESULTS The predicted core temperature and systemic oxaliplatin concentration were comparable to the values found in literature. Adequate placement of catheters, additional inflow catheters and higher flow rates reduced intraperitoneal temperature spatial variation by -1.4 °C, -2.3 °C and -1.2 °C, respectively. Flow alternations resulted in higher temperatures (up to +1.5 °C) over the peritoneal surface. Higher flow rates also reduced the spatial variation of chemotherapy concentration over the peritoneal surface resulting in a more homogeneous effective treatment dose. CONCLUSION The presented treatment planning software provides unique insights in the dynamics during HIPEC, which enables optimization of treatment-specific parameters and provides an excellent basis for HIPEC treatment planning in human applications.
Collapse
Affiliation(s)
- Daan R Löke
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Roxan F C P A Helderman
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Hans M Rodermond
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Pieter J Tanis
- Department for Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Geert J Streekstra
- Department of Biomedical Engineering and Physics, Amsterdam Movement Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicolaas A P Franken
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Arlene L Oei
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Löke DR, Helderman RFCPA, Sijbrands J, Rodermond HM, Tanis PJ, Franken NAP, Oei AL, Kok HP, Crezee J. A Four-Inflow Construction to Ensure Thermal Stability and Uniformity during Hyperthermic Intraperitoneal Chemotherapy (HIPEC) in Rats. Cancers (Basel) 2020; 12:E3516. [PMID: 33255921 PMCID: PMC7760897 DOI: 10.3390/cancers12123516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/16/2020] [Accepted: 11/25/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Hyperthermic intraperitoneal chemotherapy (HIPEC) after cytoreductive surgery (CRS) is used for treating peritoneal metastases of various origins. Present HIPEC protocols have rarely been validated for relevant parameters such as optimal agent, duration and perfusate temperature. In vitro experiments are not completely representative of clinical circumstances. Therefore, a good preclinical in vivo HIPEC model is needed in which temperature distributions can be well-controlled and are stable throughout treatments. METHODS We designed a setup able to generate and maintain a homogeneous flow during a 90-min HIPEC procedure using our in-house developed treatment planning tools and computer aided design (CAD) techniques. Twelve rats were treated with heated phosphate-buffered saline (PBS) using two catheter setups (one vs. four- inflows) and extensive thermometry. Simulated and measured thermal distribution and core temperatures were evaluated for the different setups. RESULTS Overall, the four-inflow resulted in more stable and more homogeneous thermal distributions than the one-inflow, with lower standard deviations (0.79 °C vs. 1.41 °C at the outflow, respectively) and less thermal losses. The average thermal loss was 0.4 °C lower for rats treated with the four-inflow setup. Rat core temperatures were kept stable using occasional tail cooling, and rarely exceeded 39 °C. CONCLUSION Increasing the number of inflow catheters from one to four resulted in increased flow and temperature homogeneity and stability. Tail cooling is an adequate technique to prevent rats from overheating during 90-min treatments. This validated design can improve accuracy in future in vivo experiments investigating the impact of relevant parameters on the efficacy of different HIPEC protocols.
Collapse
Affiliation(s)
- Daan R. Löke
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
| | - Roxan F. C. P. A. Helderman
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
- Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - Jan Sijbrands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
| | - Hans M. Rodermond
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
- Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - Pieter J. Tanis
- Department for Surgery, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands;
| | - Nicolaas A. P. Franken
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
- Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - Arlene L. Oei
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
- Department for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - H. Petra Kok
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (D.R.L.); (R.F.C.P.A.H.); (J.S.); (H.M.R.); (N.A.P.F.); (A.L.O.); (H.P.K.)
| |
Collapse
|
17
|
de Jong LAW, Elekonawo FMK, Lambert M, de Gooyer JM, Verheul HMW, Burger DM, de Wilt JHW, Chatelut E, Ter Heine R, de Reuver PR, Bremers AJA, van Erp NP. Wide variation in tissue, systemic, and drain fluid exposure after oxaliplatin-based HIPEC: results of the GUTOX study. Cancer Chemother Pharmacol 2020; 86:141-150. [PMID: 32594200 PMCID: PMC7338818 DOI: 10.1007/s00280-020-04107-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/24/2020] [Indexed: 01/02/2023]
Abstract
Purpose In this exploratory study, the effect of postprocedural flushing with crystalloids after oxaliplatin-based hyperthermic intraperitoneal chemotherapy (HIPEC) on platinum concentrations in peritoneal tissue, blood, and drain fluid was studied. Interpatient variability in oxaliplatin pharmacokinetics and the relation between platinum concentration in peritoneal fluid and platinum exposure in tissue and blood was explored. Methods Ten patients with peritoneal carcinomatosis of colorectal origin were treated with HIPEC including postprocedural flushing, followed by ten patients without flushing afterwards. Tissue, peritoneal fluid, blood, and drain fluid samples were collected for measurement of total and ultrafiltered platinum concentrations. Results Peritoneal tissue concentration and systemic ultrafiltered platinum exposure showed large inter individual variability, ranging from 65 to 1640 µg/g dry weight and 10.5 to 28.0 µg*h/ml, respectively. No effect of flushing was found on geometric mean platinum concentration in peritoneal tissue (348 vs. 356 µg/g dry weight), blood (14.8 vs. 18.1 µg*h/ml), or drain fluid (day 1: 7.6 vs. 7.7 µg/ml; day 2: 1.7 vs. 1.9 µg/ml). The platinum concentration in peritoneal fluid at the start of HIPEC differed twofold between patients and was positively correlated with systemic exposure (p = .04) and peak plasma concentration (p = .04). Conclusion In this exploratory study, no effect was found for postprocedural flushing on platinum concentrations in peritoneal tissue, blood, or drain fluid. BSA-based HIPEC procedure leads to large interpatient variability in platinum exposure in all compartments. The study was registered at ClinicalTrials.gov on 7 December 2017 under registration number NCT03364907.
Collapse
Affiliation(s)
- Loek A W de Jong
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Fortuné M K Elekonawo
- Department of Radiology and Nuclear Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marie Lambert
- Institut Claudius‑Regaud, IUCT‑Oncopole, and CRCT, Université de Toulouse, Inserm, 1, avenue Irène Joliot‑Curie, Toulouse, France
| | - Jan Marie de Gooyer
- Department of Radiology and Nuclear Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Etienne Chatelut
- Institut Claudius‑Regaud, IUCT‑Oncopole, and CRCT, Université de Toulouse, Inserm, 1, avenue Irène Joliot‑Curie, Toulouse, France
| | - Rob Ter Heine
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Philip R de Reuver
- Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Andre J A Bremers
- Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|