1
|
Tinsley N, O'Dwyer ST, Nagaraju R, Braun M, Mullamitha S, Kamposioras K, Marti FEM, Saunders M, Clouston H, Selvasekar C, Wild J, Wilson M, Renehan A, Aziz O, Barriuso J. Oncological Outcomes From Cytoreductive Surgery and Heated Intraperitoneal Chemotherapy for Colorectal Cancer Peritoneal Metastases. Clin Colorectal Cancer 2025; 24:166-179. [PMID: 39800608 DOI: 10.1016/j.clcc.2024.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/04/2024] [Accepted: 12/03/2024] [Indexed: 05/24/2025]
Abstract
BACKGROUND Cytoreductive surgery (CRS) is effective for colorectal cancer peritoneal metastases (CRPM) at increasing overall survival (OS) compared to systemic anticancer treatment (SACT) alone. The addition of Oxaliplatin heated intraperitoneal chemotherapy (HIPEC) has been shown in a randomized controlled trial to result in increased complications without significant OS benefit. This study evaluates outcomes for CRPM patients undergoing CRS+HIPEC with Oxaliplatin (Ox) 368mg/m2 (30 min), versus Mitomycin C (MMC) 35mg/m2 (90min). METHODS: A prospective CRPM real-world database was used to collect outcomes for patients undergoing CRS+HIPEC at a single center. OS, recurrence-free (RFS), peritoneal RFS (PeRFS) were compared amongst all patients with histologically proven CRPM, those with completeness of cytoreduction (CC) score =0/1, and those with CC score=0/1 who were SACT naïve. RESULTS: Between April 2005 and April 2021, 409 patients underwent CRS+HIPEC: 271 (66%) had MMC, 138 (34%) Ox. Of these, 395 (97%) had histologically confirmed CRPM, 336 (85%) achieved CC=0/1, 188 (47%) were SACT naïve; median OS =39.5, 44.4, and 47.2 months respectively. MMC versus Ox median OS in CC0/1=43.7 (95% CI 35.9-48.3) versus 50.1 (39.7-70.2) months, P = .28; Median OS in SACT naïve=45.7 (39.4-65.9) versus 59.9 (38.3-82.0) months, P = .31; multivariable analysis for CC0/1, SACT naïve patients showed Ox was comparable to MMC: HR=0.90, (0.64-1.27) P = .55 versus HR=0.88, (0.53-1.44) P = .60, respectively. Ox resulted in a significantly improved PeRFS in CC0/1 patients (MMC=9.0 versus Ox=12.6months, P = .01). A multivariable model for PeRFS showed a HR=0.63, (0.43-0.95), P = .03 for Ox. CONCLUSION: This study suggests a role for Ox HIPEC in CRPM which should be explored further in clinical trials.
Collapse
Affiliation(s)
- Nadina Tinsley
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Sarah T O'Dwyer
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Raghavendar Nagaraju
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Michael Braun
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Saifee Mullamitha
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Konstantinos Kamposioras
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - F E Marti Marti
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mark Saunders
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Hamish Clouston
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Chelliah Selvasekar
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Jonathan Wild
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Malcolm Wilson
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Andrew Renehan
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Omer Aziz
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom.
| | - Jorge Barriuso
- Colorectal and Peritoneal Oncology Centre (CPOC), The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
2
|
Aguirre-Allende I, Pereira-Pérez F, Manzanedo-Romero I, Fernandez-Briones P, Muñoz-Martín M, Serrano-Moral Á, Perez-Viejo E. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for colorectal peritoneal metastases: A pragmatic comparison of oncological outcomes in synchronous versus metachronous disease. Surg Oncol 2025; 58:102183. [PMID: 39778343 DOI: 10.1016/j.suronc.2024.102183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND disease burden (PCI), completeness of cytoreduction or histological features, are known to influence survival after CRS-HIPEC for colorectal peritoneal metastases (CPM). However, there is still debate about influence of CPM onset. The aim of this study is to determine the impact of CPM onset on oncological outcomes after CRS-HIPEC. METHODS all patients with CPM scheduled for CRS-HIPEC at one reference center between December 2007 and September 2022 were included. s-PM were defined as those diagnosed at primary disease treatment; m-PM were considered those diagnosed during follow-up. Survival outcomes and recurrence rates were compared using a pragmatic analysis. RESULTS 125 patients with s-CPM and 170 patients with m-CPM were analyzed. Median follow-up was 58.6 and 50.6 months in s-CPM and m-CPM groups(p = 0.11). Complete cytoreduction (CCS-0/-1) rates were comparable: 84 % s-CPM vs. 88.2 % m-CPM(p = 0.190). Overall survival (OS) was significantly shorter in s-CPM: 24.7 vs. 46.6 months (p = 0.024). Conversely, median disease-free survival was similar in both groups, 10 months vs. 11 months(p = 0.155). Patients in the s-CPM group presented more pN+(p = 0.001), higher histologic grade(p = 0.007) and PCI(p = 0.04), and higher rate of concurrent liver metastases(p = 0.004). RAS/BRAF gene mutations and microsatellite instability did not differ significantly. Perioperative chemotherapy regimens and tolerance were also similar. CONCLUSIONS despite s-CPM being associated with impaired OS after CRS-HIPEC, the onset of PM was not found to be an independent determinant for survival. High-risk molecular and histological features strongly influence oncological outcomes after CRS-HIPEC. This is valuable data that could aid in preoperative patient selection process for CRS-HIPEC.
Collapse
Affiliation(s)
- Ignacio Aguirre-Allende
- Hepatobiliary and Peritoneal Surface Malignancies Unit. General and Digestive Surgery Department, Donostia University Hospital-IHO Donostialdea, IIS Biodonostia, Spain.
| | - Fernando Pereira-Pérez
- Peritoneal Surface Malignancies Unit, General and Digestive Surgery Department, Fuenlabrada University Hospital, Madrid, Spain
| | - Israel Manzanedo-Romero
- Peritoneal Surface Malignancies Unit, General and Digestive Surgery Department, Fuenlabrada University Hospital, Madrid, Spain
| | - Paula Fernandez-Briones
- Peritoneal Surface Malignancies Unit, General and Digestive Surgery Department, Fuenlabrada University Hospital, Madrid, Spain
| | - María Muñoz-Martín
- Peritoneal Surface Malignancies Unit, General and Digestive Surgery Department, Fuenlabrada University Hospital, Madrid, Spain
| | - Ángel Serrano-Moral
- Peritoneal Surface Malignancies Unit, General and Digestive Surgery Department, Fuenlabrada University Hospital, Madrid, Spain
| | - Estibalitz Perez-Viejo
- Peritoneal Surface Malignancies Unit, General and Digestive Surgery Department, Fuenlabrada University Hospital, Madrid, Spain
| |
Collapse
|
3
|
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
|
4
|
Kusamura S, Bhatt A, van Der Speeten K, Kepenekian V, Hübner M, Eveno C, de Hingh I, Delhorme J, Taibi A, Villeneuve L, Dico RL, Moran B, Govaerts K, Zivanovic O, Brennan D, Nadeau C, Van Driel W, Bakrin N, Piso P, Verwaal VJ, González‐Moreno S, Alyami M, Sgarbura O, Rau B, Deraco M, Glehen O. Review of 2022 PSOGI/RENAPE Consensus on HIPEC. J Surg Oncol 2024; 130:1290-1298. [PMID: 39285659 PMCID: PMC11826010 DOI: 10.1002/jso.27885] [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] [Accepted: 08/26/2024] [Indexed: 02/16/2025]
Abstract
The 2022 PSOGI (Peritoneal Surface Oncology Group International) and RENAPE (French Network for Rare Peritoneal Malignancies) consensus on hyperthermic intraperitoneal chemotherapy (HIPEC) was a comprehensive effort aimed at standardizing treatment protocols for various peritoneal malignancies. This initiative is critical due to the wide range of technical variations in HIPEC procedures and the resulting need for standardization to ensure consistent and effective patient care and meaningful audit of multicenter data.
Collapse
Affiliation(s)
- Shigeki Kusamura
- PSM Unit, Department of Surgical OncologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Aditi Bhatt
- Department of Surgical OncologyKD HospitalAhmedabadIndia
| | - Kurt van Der Speeten
- Department of Abdominal and Oncological SurgeryZiekenhuis Oost Limburg (ZOL)GenkBelgium
| | - Vahan Kepenekian
- Department of Oncological SurgeryHôpital Lyon Sud, Hospices Civils de LyonLyonFrance
- CICLYUniversité Claude Bernard Lyon 1LyonFrance
| | - Martin Hübner
- Department of Visceral SurgeryLausanne University Hospital (CHUV), University of Lausanne (UNIL)LausanneSwitzerland
| | - Clarisse Eveno
- Department of Digestive and Oncological SurgeryUniversity Lille, Claude Huriez University HospitalLilleFrance
| | - Ignace de Hingh
- Department of SurgeryCatharina Cancer InstituteEindhovenThe Netherlands
| | - Jean‐Baptiste Delhorme
- Department of General and Digestive SurgeryHautepierre Hospital, Strasbourg University HospitalStrasbourgFrance
| | - Abdelkader Taibi
- Department of Digestive SurgeryDupuytren University HospitalLimogesFrance
- XLIM, UMR 7252CNRSLimogesFrance
| | - Laurent Villeneuve
- Hospices Civils de Lyon, Hôpital Lyon Sud, Service de Recherche et d'Epidémiologie CliniquesPierre‐BéniteFrance
| | - Rea Lo Dico
- Department of General Surgery, Emergency, and New TechnologiesSan Camillo Forlanini HospitalRomeItaly
| | - Brendan Moran
- Peritoneal Malignancy UnitBasingstoke HospitalBasingstokeUK
| | - Kim Govaerts
- Department of Abdominal and Oncological SurgeryZiekenhuis Oost Limburg (ZOL)GenkBelgium
| | - Oliver Zivanovic
- Department of Gynecological OncologyHeidelberg Medizinische KlinikHeidelbergGermany
| | - Donal Brennan
- UCD Gynaecological Oncology GroupUCD School of Medicine, Mater Misericordiae University HospitalDublinIreland
| | - Cedric Nadeau
- Department of Gynecological OncologyClinique du Fief de GrimoirePoitiersCedexFrance
| | - Willemien Van Driel
- Department of Gynecological OncologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Naoual Bakrin
- Department of Surgical OncologyHôpita Lyon Sud, Hospices Civils de LyonLyonFrance
| | - Pompiliu Piso
- Department of General and Visceral SurgeryHospital Barmherzige BrüderRegensburgGermany
| | - Victor J. Verwaal
- Peritoneal Surface Malignancy and HIPEC Skåne University HospitalLunds University SwedenLundsSweden
| | | | - Mohammad Alyami
- Department of General Surgery and Surgical OncologyKing Khalid HospitalNajranSaudi Arabia
| | - Olivia Sgarbura
- Department of Surgical Oncology, Montpellier Cancer InstituteUniversity of MontpellierMontpellierFrance
| | - Beate Rau
- Chirurgische KlinikCampus Charité Mitte, Charité‐UniversitätsmedizinBerlinGermany
| | - Marcello Deraco
- PSM Unit, Department of Surgical OncologyFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Olivier Glehen
- Department of Oncological SurgeryHôpital Lyon Sud, Hospices Civils de LyonLyonFrance
- CICLYUniversité Claude Bernard Lyon 1LyonFrance
| |
Collapse
|
5
|
Moldovan B, Adam II, Pisică RM, Untaru V, Stoica D, Șpac A, Modrigan I, Ursu M, Jupoiu L, Frâncu A, Pescaru F, Hubeishie A, Zolog A, Vecerzan L. Takotsubo Cardiomyopathy After Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for a Recurrent Colon Cancer: A Life-Threatening Complication. Diagnostics (Basel) 2024; 14:2402. [PMID: 39518369 PMCID: PMC11545001 DOI: 10.3390/diagnostics14212402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
(1) Background: Takotsubo cardiomyopathy, or stress cardiomyopathy, is an acute heart failure condition with transient left ventricular (LV) motion abnormalities but no significant coronary artery obstruction. It mimics acute coronary syndrome (ACS), with symptoms like chest pain, dyspnea, and ECG changes. (2) Case Report: We present the case of a 44-year-old female with relapsed colon cancer and peritoneal carcinomatosis. After undergoing cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC), she experienced cardiac arrest from ventricular fibrillation 18 h postoperatively. Echocardiography revealed a reduced LV ejection fraction (20%) and apical akinesia, suggesting a Takotsubo Cardiomyopathy. Intensive resuscitation and inotropic support led to gradual recovery. Coronary angiography confirmed no coronary artery obstruction. (3) Discussion: This case highlights TTS as a rare but severe complication following major oncological surgeries, possibly triggered by both physical and emotional stressors. TTS should be considered in the differential diagnosis of perioperative cardiac events in cancer patients. (4) Conclusions: Prompt recognition and management of TTS in the perioperative period are crucial to improving outcomes, especially in high-risk oncological patients undergoing extensive surgeries.
Collapse
Affiliation(s)
- Bogdan Moldovan
- General Surgery Department, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (I.-I.A.); (R.-M.P.)
| | - Iris-Iuliana Adam
- General Surgery Department, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (I.-I.A.); (R.-M.P.)
- Faculty of Medicine, “Lucian Blaga” University, 550169 Sibiu, Romania;
| | - Radu-Mihai Pisică
- General Surgery Department, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (I.-I.A.); (R.-M.P.)
| | - Vlad Untaru
- Anesthesia and Intensive Care Unit, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (V.U.); (D.S.); (A.Ș.)
| | - Doly Stoica
- Anesthesia and Intensive Care Unit, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (V.U.); (D.S.); (A.Ș.)
| | - Alexandra Șpac
- Anesthesia and Intensive Care Unit, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (V.U.); (D.S.); (A.Ș.)
| | - Irina Modrigan
- Anesthesia and Intensive Care Unit, Sanador Hospital, 011031 Bucharest, Romania;
| | - Mihai Ursu
- Cardiology Department, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (M.U.); (L.J.); (A.F.)
| | - Liliana Jupoiu
- Cardiology Department, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (M.U.); (L.J.); (A.F.)
| | - Adina Frâncu
- Cardiology Department, ‘St. Constantin’ Hospital, 500299 Brasov, Romania; (M.U.); (L.J.); (A.F.)
| | | | - Amir Hubeishie
- Oncology Department, Timiş County Emergency Clinical Hospital, 300723 Timișoara, Romania;
- Oncology Department, Oncohelp Medical Center, 300425 Timișoara, Romania
| | - Adriana Zolog
- Pathology Department, Regina Maria Hospital, 400500 Cluj-Napoca, Romania;
| | - Liliana Vecerzan
- Faculty of Medicine, “Lucian Blaga” University, 550169 Sibiu, Romania;
- Oncology Department, ‘Dr. Alexandru Augustin’ Military Hospital, 550024 Sibiu, Romania
| |
Collapse
|
6
|
Jeong MH, Kang SJ, Park SY, Kwak SG, Seo AN, Park S, Park JS, Kim HJ, Choi GS. Comparison of EPIC Versus HIPEC in the Treatment of Colorectal Peritoneal Metastases and Appendix Tumors Using Inverse Probability of Treatment Weighting. Ann Surg Oncol 2024; 31:7111-7121. [PMID: 39073661 DOI: 10.1245/s10434-024-15674-2] [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: 12/27/2023] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The selection of hyperthermic intraperitoneal chemotherapy (HIPEC) or early postoperative intraperitoneal chemotherapy (EPIC) for peritoneal metastases from colorectal cancer or appendiceal neoplasms following cytoreductive surgery (CRS) depends on the surgeon's discretion. This study was designed to compare postoperative and oncologic outcomes of HIPEC and EPIC using inverse probability of treatment weighting (IPTW). METHODS This study included 175 patients who received HIPEC or EPIC following CRS at a single tertiary university hospital between December 1999 and December 2020. Inverse probability of treatment weighting analysis was performed to control for pretreatment characteristics between the two groups. Multivariate analysis was performed to determine factors associated with postoperative and survival outcomes. RESULTS After IPTW, no significant differences in baseline demographics and tumor characteristics were observed between the two groups. The HIPEC group had a significantly longer operation time than the EPIC group. The EPIC group showed a significantly higher postoperative mortality rate than the HIPEC group. Operation time (odds ratio [OR] 1.01; 95% confidence interval [CI] 1.01-1.02; p < 0.001), bowel anastomosis (OR 7.25; 95% CI 1.16-45.2; p = 0.034), neoadjuvant chemotherapy (OR 7.62; 95% CI 1.85-31.4; p = 0.005), and EPIC (OR 8.76; 95% CI 2.16-35.5; p = 0.002) were independent risk factors for major surgical complications. No association was observed between intraperitoneal chemotherapy type and major hematologic toxicity, overall survival, progression-free survival, or peritoneal progression-free survival. CONCLUSIONS EPIC was a risk factor for major surgical complications. Survival outcomes were similar between the two types of intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Min Hye Jeong
- Colorectal Cancer Center, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
| | - Su Jin Kang
- Colorectal Cancer Center, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
| | - Soo Yeun Park
- Colorectal Cancer Center, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea.
| | - Sang Gyu Kwak
- Department of Medical Statistics, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea.
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Suehyun Park
- Department of Surgery, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jun Seok Park
- Colorectal Cancer Center, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
| | - Hye Jin Kim
- Colorectal Cancer Center, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
| | - Gyu-Seog Choi
- Colorectal Cancer Center, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
7
|
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
|
8
|
Varinelli L, Battistessa D, Guaglio M, Zanutto S, Illescas O, Lorenc EJ, Pisati F, Kusamura S, Cattaneo L, Sabella G, Milione M, Perbellini A, Noci S, Paolino C, Kuhn E, Galassi M, Cavalleri T, Deraco M, Gariboldi M, Baratti D. Colorectal carcinoma peritoneal metastases-derived organoids: results and perspective of a model for tailoring hyperthermic intraperitoneal chemotherapy from bench-to-bedside. J Exp Clin Cancer Res 2024; 43:132. [PMID: 38698446 PMCID: PMC11064374 DOI: 10.1186/s13046-024-03052-5] [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: 01/04/2024] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Peritoneal metastases from colorectal cancer (CRCPM) are related to poor prognosis. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) have been reported to improve survival, but peritoneal recurrence rates are still high and there is no consensus on the drug of choice for HIPEC. The aim of this study was to use patient derived organoids (PDO) to build a relevant CRCPM model to improve HIPEC efficacy in a comprehensive bench-to-bedside strategy. METHODS Oxaliplatin (L-OHP), cisplatin (CDDP), mitomycin-c (MMC) and doxorubicin (DOX) were used to mimic HIPEC on twelve PDO lines derived from twelve CRCPM patients, using clinically relevant concentrations. After chemotherapeutic interventions, cell viability was assessed with a luminescent assay, and the obtained dose-response curves were used to determine the half-maximal inhibitory concentrations. Also, induction of apoptosis by different HIPEC interventions on PDOs was studied by evaluating CASPASE3 cleavage. RESULTS Response to drug treatments varied considerably among PDOs. The two schemes with better response at clinically relevant concentrations included MMC alone or combined with CDDP. L-OHP showed relative efficacy only when administered at low concentrations over a long perfusion period. PDOs showed that the short course/high dose L-OHP scheme did not appear to be an effective choice for HIPEC in CRCPM. HIPEC administered under hyperthermia conditions enhanced the effect of chemotherapy drugs against cancer cells, affecting PDO viability and apoptosis. Finally, PDO co-cultured with cancer-associated fibroblast impacted HIPEC treatments by increasing PDO viability and reducing CASPASES activity. CONCLUSIONS Our study suggests that PDOs could be a reliable in vitro model to evaluate HIPEC schemes at individual-patient level and to develop more effective treatment strategies for CRCPM.
Collapse
Affiliation(s)
- Luca Varinelli
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Davide Battistessa
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Marcello Guaglio
- Peritoneal Surface Malignancies Unit, Colorectal Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Susanna Zanutto
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Oscar Illescas
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Ewelina J Lorenc
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Federica Pisati
- Cogentech Ltd. Benefit Corporation With a Sole Shareholder, Via Adamello 16, Milan, 20139, Italy
| | - Shigeki Kusamura
- Peritoneal Surface Malignancies Unit, Colorectal Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Laura Cattaneo
- Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via G. Venezian 1, Milan, 20133, Italy
| | - Giovanna Sabella
- Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via G. Venezian 1, Milan, 20133, Italy
| | - Massimo Milione
- Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via G. Venezian 1, Milan, 20133, Italy
| | - Alessia Perbellini
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Sara Noci
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Cinzia Paolino
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Elisabetta Kuhn
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, 20122, Italy
- Pathology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Margherita Galassi
- Centrale Produzione Farmaci, Hospital Pharmacy, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via G. Venezian 1, Milan, 20133, Italy
| | - Tommaso Cavalleri
- Peritoneal Surface Malignancies Unit, Colorectal Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Marcello Deraco
- Peritoneal Surface Malignancies Unit, Colorectal Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy.
| | - Manuela Gariboldi
- Department of Experimental Oncology, Molecular Epigenomics Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| | - Dario Baratti
- Peritoneal Surface Malignancies Unit, Colorectal Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, Milan, 20133, Italy
| |
Collapse
|
9
|
Pei B, Zhu J, Lai L. Comment on 'Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy versus R0 resection for resectable colorectal cancer with peritoneal metastases and low peritoneal cancer index scores: A collaborative observational study from Korea and Japan'. Int J Surg 2024; 110:643-644. [PMID: 38016290 PMCID: PMC10871642 DOI: 10.1097/js9.0000000000000932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023]
Affiliation(s)
- Bo Pei
- Department of Oncology, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University
| | - Jia Zhu
- Department of Clinical Medicine, Hubei Enshi College, Enshi, People’s Republic of China
| | - Lin Lai
- Department of Oncology, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University
| |
Collapse
|
10
|
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
|
11
|
Kitaguchi D, Park EJ, Baik SH, Sasaki S, Tsukada Y, Ito M. Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy versus R0 resection for resectable colorectal cancer with peritoneal metastases and low peritoneal cancer index scores: a collaborative observational study from Korea and Japan. Int J Surg 2024; 110:45-52. [PMID: 37800569 PMCID: PMC10793799 DOI: 10.1097/js9.0000000000000809] [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: 07/19/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND The benefits of hyperthermic intraperitoneal chemotherapy (HIPEC) after cytoreductive surgery (CRS) for colorectal cancer with peritoneal metastasis (CPM) remain controversial. R0 resection without peritoneal stripping might be as effective as CRS plus HIPEC. The authors aimed to compare the long-term oncological outcomes of patients with CPM and peritoneal cancer index (PCI) scores less than or equal to 6 who underwent R0 resection in Japan with those who underwent CRS plus HIPEC in Korea. MATERIALS AND METHODS This international, retrospective cohort study was conducted in Korea and Japan using a prospectively collected clinical database. Patients who underwent surgery from July 2014 to December 2021 for CPM with a PCI score of less than or equal to 6 and completeness of the cytoreduction score-0 were included. The primary outcome was relapse-free survival (RFS), and the secondary outcomes were overall survival, peritoneal RFS (PRFS), and postoperative outcomes. RESULTS The 3-year RFS was significantly longer in the CRS+HIPEC group than in the R0 resection group: 35.9% versus 6.9% ( P <0.001); 31.0% versus 6.7% ( P =0.040) after propensity score matching. The median PRFS was significantly longer in the CRS+HIPEC group than in the R0 resection group: 24.5 months versus 17.2 months ( P =0.017). The 3-year overall survival and postoperative complications did not significantly differ between the two groups. CONCLUSIONS RFS and PRFS rates were significantly prolonged after CRS plus HIPEC, whereas postoperative complications and length of hospital stay were not increased. Therefore, curative CRS plus HIPEC may be considered a treatment strategy for selected patients with resectable CPM and low PCI scores.
Collapse
Affiliation(s)
- Daichi Kitaguchi
- Department of Colorectal Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Eun Jung Park
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine
| | - Seung Hyuk Baik
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine
- Mirae Medical Check-up Hospital, Seoul, Korea
| | - Shoma Sasaki
- Department of Colorectal Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Yuichiro Tsukada
- Department of Colorectal Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Masaaki Ito
- Department of Colorectal Surgery, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
12
|
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
|
13
|
Yu P, Huang X, Huang L, Dai G, Xu Q, Fang J, Ye Z, Chai T, Du Y. Hyperthermic intraperitoneal chemotherapy (HIPEC) plus systemic chemotherapy versus systemic chemotherapy alone in locally advanced gastric cancer after D2 radical resection: a randomized-controlled study. J Cancer Res Clin Oncol 2023; 149:11491-11498. [PMID: 37392201 PMCID: PMC10465671 DOI: 10.1007/s00432-023-05019-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND Currently, there is a lack of an effective strategy for the prevention of peritoneal metastasis (PM) from locally advanced gastric cancer (AGC). This randomized-controlled study aimed to evaluate the outcome of D2 radical resection with hyperthermic intraperitoneal chemotherapy (HIPEC) plus systemic chemotherapy versus systemic chemotherapy alone in locally AGC patients. METHODS All enrolled patients were randomly assigned to receive HIPEC plus systemic chemotherapy (HIPEC group) or systemic chemotherapy alone (non-HIPEC group) after radical gastrectomy. HIPEC was performed intraperitoneally with cisplatin (40 mg/m2) within 72 h after surgery, while systemic chemotherapy based on the SOX regimen (S-1 combined with oxaliplatin) was administered 4-6 weeks after radical surgery. Patterns of recurrence, adverse events, 3-year disease-free survival (DFS), and overall survival (OS) were analyzed. RESULTS A total of 134 patients were enrolled in the present study. The 3-year DFS rate was 73.8% in the HIPEC group, which was significantly higher than that in the non-HIPEC group (61.2%, P = 0.031). The 3-year OS rate was 73.9% in the HIPEC group and 77.6% in the non-HIPEC group, with no significant difference (P = 0.737). PM was the most common distant metastasis in both groups. The occurrence rate of PM in the HIPEC group was statistically lower than that in the non-HIPEC group (20.9% vs. 40.3%, P = 0.015). Grade 3 or 4 adverse events occurred in 19 (14.2%) patients, and there was no significant difference between the two groups. CONCLUSION Radical surgery followed by HIPEC combined with systemic chemotherapy is a safe and feasible strategy for locally AGC patients and could effectively improve DFS and reduce the occurrence of PM. However, more prospective randomized studies with a large sample size are warranted. TRIAL REGISTRATION This study was registered with www.medresman.org.cn as ChiCTR2200055966 on 10/12/2016.
Collapse
Affiliation(s)
- Pengfei Yu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xingmao Huang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Ling Huang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Gaiguo Dai
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qi Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jingquan Fang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zeyao Ye
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Tengjiao Chai
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yian Du
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
14
|
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
|
15
|
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] [Download PDF] [Figures] [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
|
16
|
Park SY, Park JS, Kim HJ, Kim JG, Kang BW, Baek JH, Kim HR, Kim CH, Kim YJ, Choi GS. Oncological impact of intraperitoneal chemotherapy after cytoreductive surgery for patients with colorectal peritoneal metastasis: A bi-institutional retrospective analysis. J Surg Oncol 2023; 127:668-677. [PMID: 36515216 DOI: 10.1002/jso.27171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/28/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES There is a paucity of evidence on the value of intraperitoneal chemotherapy (IPC) following cytoreductive surgery (CRS) for colorectal peritoneal metastasis. This study aimed to evaluate the association between mitomycin C-IPC and survival outcomes following CRS. METHODS The institutional databases of two tertiary hospitals were reviewed to identify patients who underwent CRS for colorectal peritoneal metastasis. The outcomes of patients who underwent CRS without IPC were compared with those of patients who underwent CRS plus early postoperative intraperitoneal chemotherapy (EPIC) or CRS plus hyperthermic intraperitoneal chemotherapy (HIPEC). The primary endpoints were cancer-specific survival (CSS), progression-free survival (PFS), and peritoneal PFS (P-PFS). RESULTS In 149 patients with peritoneal metastasis alone, EPIC and HIPEC use was significantly associated with better CSS, PFS, and P-PFS in the multivariate analysis. CSS was also significantly associated with perioperative systemic chemotherapy. Among 42 patients with both peritoneal and extraperitoneal metastases, CSS was independently related to the completeness of cytoreduction score, location of extraperitoneal metastasis, and grade 3-4 complications. CONCLUSIONS Mitomycin C-IPC after CRS was associated with better survival outcomes than CRS alone in patients with resectable peritoneal metastasis of colorectal cancer. This study found that IPC had beneficial effects regarding P-PFS in patients with both peritoneal and extraperitoneal metastases.
Collapse
Affiliation(s)
- Soo Yeun Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Seok Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hye Jin Kim
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jong Gwang Kim
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Ho Baek
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyeong Rok Kim
- Department of Surgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun, Republic of Korea
| | - Chang Hyun Kim
- Department of Surgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun, Republic of Korea
| | - Young Jin Kim
- Department of Surgery, St. Carollo General Hospital, Suncheon-si, Republic of Korea
| | - Gyu-Seog Choi
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
17
|
Fanget F, Kefleyesus A, Peron J, Bonnefoy I, Villeneuve L, Passot G, Rousset P, You B, Benzerdjeb N, Glehen O, Kepenekian V. Comparison of Neoadjuvant Systemic Chemotherapy Protocols for the Curative-Intent Management of Peritoneal Metastases from Colorectal Cancer, Regarding Morphological Response, Pathological Response, and Long-Term Outcomes: A Retrospective Study. Ann Surg Oncol 2023; 30:3304-3315. [PMID: 36729351 DOI: 10.1245/s10434-023-13150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/12/2023] [Indexed: 02/03/2023]
Abstract
BACKGROUND Selected patients with colorectal cancer peritoneal metastases (CRPM) could be offered a curative-intent strategy based on complete cytoreductive surgery (CRS), potentially combined with hyperthermic intraperitoneal chemotherapy (HIPEC) and perioperative systemic chemotherapy. The impact of different neoadjuvant systemic chemotherapy (NACT) regimens remains unclear due to a lack of comparative data. METHODS Consecutive CRPM patients from a monocentric database who were treated with complete CRS after single-line NACT were included in this study. Chemotherapy regimens were tailored as a doublet drug (FOLFOX/FOLFIRI) with/without targeted therapy (anti-epidermal growth factor receptor/bevacizumab) and triplet-drug combination (FOLFIRINOX). Morphological response (MR) was assessed using the Response Evaluation Criteria in Solid Tumors criteria, and pathological response (PR) was assessed using the Peritoneal Regression Grading Score (PRGS). Long-term oncologic outcomes were compared. RESULTS The cohort comprised 388 patients, including 127, 202, and 59 patients in the doublet, doublet + targeted, and triplet groups, respectively. MR rates were higher in the triplet (68.0%) and doublet + targeted groups (64.2%) when compared with the doublet group (42.4%, p = 0.003). Complete and major PRs were observed in 13.6% and 32.0% of patients, respectively. Higher MR rates were observed after doublet + targeted or triplet regimens, while no difference was observed for PR rates. In multivariate analysis, FOLFIRINOX was independently associated with better overall survival (hazard ratio 0.49, 95% confidence interval 0.25-0.96; p = 0.037). FOLFIRINOX also resulted in a higher rate of severe postoperative complications. CONCLUSIONS In this retrospective study, a FOLFIRINOX regimen as NACT seemed to result in better long-term outcomes for CRPM patients after complete CRS/HIPEC, although with higher morbidity. Prospective studies are needed, including groups without NACT and those with FOLFIRINOX + bevacizumab.
Collapse
Affiliation(s)
- Florian Fanget
- Surgical Oncology Department, Service de Chirurgie Digestive et Oncologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Amaniel Kefleyesus
- Surgical Oncology Department, Service de Chirurgie Digestive et Oncologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,Department of Visceral Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
| | - Julien Peron
- Medical Oncology Department, Laboratoire de Biométrie et Biologie Evolutive, Université Claude Bernard Lyon I (UCBL1), Hôpital Lyon Sud, Hospices Civils de Lyon, Equipe Biostatistique-Santé, Lyon, France
| | - Isabelle Bonnefoy
- EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Laurent Villeneuve
- EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Guillaume Passot
- Surgical Oncology Department, Service de Chirurgie Digestive et Oncologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Pascal Rousset
- EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France.,Department of Radiology, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Bénite, France
| | - Benoit You
- EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France.,Medical Oncology Department, Laboratoire de Biométrie et Biologie Evolutive, Université Claude Bernard Lyon I (UCBL1), Hôpital Lyon Sud, Hospices Civils de Lyon, Equipe Biostatistique-Santé, Lyon, France
| | - Nazim Benzerdjeb
- EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France.,Department of Pathology, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Bénite, France
| | - Olivier Glehen
- Surgical Oncology Department, Service de Chirurgie Digestive et Oncologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.,EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Vahan Kepenekian
- Surgical Oncology Department, Service de Chirurgie Digestive et Oncologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France. .,EA3738 CICLY, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France.
| |
Collapse
|
18
|
Wu L, Zhu W, Meng J, Wu J, Li L, Fang C, Zhang H. A single-center randomized controlled study of intraperitoneal hyperthermic chemoperfusion in combination of recombinant human tumor necrosis factor (TNF) in treatment of malignant ascites caused by advanced abdominal cancers. Medicine (Baltimore) 2022; 101:e31883. [PMID: 36482606 PMCID: PMC9726325 DOI: 10.1097/md.0000000000031883] [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] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Malignant peritoneal effusion is a common complication of advanced malignancies, which has a poor prognosis for patients. Hyperthermic intraperitoneal chemotherapy (HIPEC) has been widely used in the treatment of advanced gynecological tumors, especially ovarian cancer (OC). Relative studies have indicated that HIPEC allows for direct exposure of tumor cells to high peritoneal concentrations of cytotoxic drugs without increasing systemic toxicity compared with intravenous treatment. Recombinant human tumor necrosis factor for injection (rmhTNF-NC) is a safely tolerated immunotherapeutic drug that has becoming a mainstay of treatment for malignant effusions. Currently, a prospective study is required to determining the efficacy of rmhTNF-NC plus cisplatin for the treatment of malignant peritoneal effusion for OC. METHODS Design and setting: This is a single-center, open trial will be performed in Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine. PARTICIPANTS Eligible patients will be those with advanced gynecologic cancers and who would be suitable for HIPEC. INTERVENTION AND CONTROL HIPEC with cisplatin and intraperitoneal perfusion with rmhTNF-NC. COINTERVENTIONS Further chemotherapy will be offered to patients as per current practice.OutcomesPilot study: Patients and clinicians' acceptability of the trial to assist in optimization of recruitment.Primary outcome: One-year overall survival (OS).Secondary outcomes: Progression-free survival (PFS), adverse events.Follow-up: One-year follow-up for OS.Sample size: Twenty patients to demonstrate therapeutic effect of peritoneal effusion caused by OC. DISCUSSION This trial will determine the effectiveness of HIPEC with cisplatin and intraperitoneal perfusion with rmhTNF-NC for advanced gynecologic cancers, and guide the optimal treatment for these patients.
Collapse
Affiliation(s)
- Lexia Wu
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Wanshan Zhu
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Jincheng Meng
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Jiaming Wu
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Luzhen Li
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Cantu Fang
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Huatang Zhang
- Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan, China
- *Correspondence: Huatang Zhang, Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, Zhongshan 528400, China (e-mail: )
| |
Collapse
|
19
|
Park EJ, Baik SH. Recent Advance in the Surgical Treatment of Metastatic Colorectal Cancer-An English Version. J Anus Rectum Colon 2022; 6:213-220. [PMID: 36348943 PMCID: PMC9613413 DOI: 10.23922/jarc.2022-048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/04/2022] [Indexed: 11/06/2022] Open
Abstract
Stage IV colorectal cancer (CRC) has heterogeneous characteristics in tumor extent and biology. The overall survival of patients with metastatic CRC has improved with the development of multimodal treatments and new chemotherapeutic drugs. Resection of metastatic CRC is performed for liver, lung, or peritoneal metastases. Conversion surgeries to resect oligometastatic lesions have been developed with tumor regression using chemotherapeutic agents. Two-stage hepatectomy has extended the surgical indications for patients with metastatic CRC. Synchronous liver and primary tumor resection can be considered in patients with adequate conditions. Local ablation with radiotherapy can be used to treat lung metastasis. In the treatment of patients with CRC with peritoneal metastasis, cytoreductive surgery with hyperthermic intraperitoneal chemotherapy can be considered. Surgical treatments should be performed in patients with symptomatic primary tumors with unresectable metastasis. However, primary tumor resection in patients with asymptomatic CRC with synchronous, unresectable metastases did not show overall survival benefits in recent studies. Therefore, the treatment of metastatic CRC is challenging due to the variable tumor extent and heterogenous characteristics. Tailored surgical treatments and multidisciplinary approaches may improve survival and the quality of life in patients with metastatic CRC.
Collapse
Affiliation(s)
- Eun Jung Park
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine
| | - Seung Hyuk Baik
- Division of Colon and Rectal Surgery, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine
| |
Collapse
|
20
|
Ammerata G, Filippo R, Laface C, Memeo R, Solaini L, Cavaliere D, Navarra G, Ranieri G, Currò G, Ammendola M. Hyperthermic intraperitoneal chemotherapy and colorectal cancer: From physiology to surgery. World J Clin Cases 2022; 10:10852-10861. [PMID: 36338235 PMCID: PMC9631165 DOI: 10.12998/wjcc.v10.i30.10852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/23/2022] [Accepted: 08/13/2022] [Indexed: 02/05/2023] Open
Abstract
The pursuit of this paper is to collect principal reviews and systematic reviews about hyperthermic intraperitoneal chemotherapy (HIPEC) and cytoreductive surgery (CRS) used in colorectal cancer (CRC). We focus on principal biological aspects of CRC, hyperthermia effects, and surgical procedures. We searched PubMed/MEDLINE for the principal reviews and systematic reviews published from 2010 to 2021 regarding the bimodal treatment (CRS + HIPEC) against local and advanced CRC. In the literature, from several studies, it seems that the efficacy of bimodal treatment with an accurate CRS can extend overall survival. Despite these studies, there are not still any straight guidelines more detailed and scheduled about the use of combined treatment in patients with CRC. Even if the concept is still not very clear and shared, after a careful evaluation of the published data, and after some technical and pathophysiological descriptions, we concluded that it is possible to improve the overall survival and quality of life and to reduce the tumor relapse in patients affected by locally advanced (pT4) CRC with peritoneal metastases.
Collapse
Affiliation(s)
- Giorgio Ammerata
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| | - Rosalinda Filippo
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| | - Carmelo Laface
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre “Giovanni Paolo II”, Bari 70124, Italy
| | - Riccardo Memeo
- Hepato-Biliary and Pancreatic Surgical Unit, “F. Miulli” Hospital, Acquaviva delle Fonti, Bari 70124, Italy
| | - Leonardo Solaini
- Department of Medical and Surgical Sciences, University of Bologna, Forlì 40126, Italy
| | - Davide Cavaliere
- Department of General and Oncologic Surgery, Morgagni-Pierantoni Hospital, Forlì 47121, Italy
| | - Giuseppe Navarra
- Department of Human Pathology of Adult and Evolutive Age, Surgical Oncology Division, “G. Martino” Hospital, University of Messina, Messina 98122, Italy
| | - Girolamo Ranieri
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre “Giovanni Paolo II”, Bari 70124, Italy
| | - Giuseppe Currò
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| | - Michele Ammendola
- Science of Health Department, Digestive Surgery Unit, University “Magna Graecia” Medical School, Catanzaro 88100, Italy
| |
Collapse
|
21
|
Park EJ, Baik SH. Surgical treatment for metastatic colorectal cancer. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2022. [DOI: 10.5124/jkma.2022.65.9.568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Stage IV colorectal cancer (CRC) exhibits heterogeneous characteristics in tumor extent and biology. The overall survival of patients with metastatic CRC has improved with the development of multimodal treatments and new chemotherapeutic drugs.Current Concepts: Resection of metastatic CRC is performed for liver, lung, or peritoneal metastases. Conversion surgeries to resect oligometastatic lesions have been developed with tumor regression using chemotherapeutic agents. Two-stage hepatectomy has extended the surgical indications for patients with metastatic CRC. Synchronous liver and primary tumor resection can be considered in patients with adequate conditions. Local ablation with radiotherapy can be used to treat lung metastasis. Meanwhile, for treating patients with CRC with peritoneal metastasis, cytoreductive surgery with hyperthermic intraperitoneal chemotherapy can be considered. Surgical treatments should be performed in patients with symptomatic primary tumors with unresectable metastasis. However, in recent studies, primary tumor resection in patients with asymptomatic CRC with synchronous, unresectable metastases did not show overall survival benefits.Discussion and Conclusion: The treatment of metastatic CRC is challenging because of the variable tumor extent and heterogenous characteristics. Tailored surgical treatments and multidisciplinary approaches may improve the survival and quality of life of patients with metastatic CRC.
Collapse
|
22
|
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]
|
23
|
Pacelli F, Gerardi C, Rulli E, Abatini C, Rotolo S, Garattini S, Melotti G, Torri V, Galli F, Rulli E, Di Giorgio A. Prophylactic surgery plus hyperthermic intraperitoneal chemotherapy (HIPEC CO2) versus standard surgery in colorectal carcinoma at high risk of peritoneal carcinomatosis: short-term and long-term outcomes from the CHECK study - protocol for a randomised, multicentre, phase 3 trial. BMJ Open 2022; 12:e051324. [PMID: 35914916 PMCID: PMC9345052 DOI: 10.1136/bmjopen-2021-051324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Up to one-fifth of patients with colorectal cancer will develop peritoneal metastases, frequently without other districts' involvement. Despite the recent unsuccesses of hyperthermic intraperitoneal chemotherapy (HIPEC) for colorectal cancer peritoneal metastases treatment, the rationale in the prophylactic setting remains strong. Several clinical and pharmacokinetic data suggest that the efficacy of intraperitoneal chemotherapy is highest when the disease is microscopic. However, robust evidence demonstrating whether the addition of HIPEC for high-risk colorectal cancers offers better control of local recurrence is lacking. METHODS AND ANALYSIS This is a multicentre randomised phase 3 trial comparing prophylactic surgery plus HIPEC CO2 with mitomycin, over standard surgical excision in patients with colorectal cancer at high risk of peritoneal carcinomatosis; 388 patients will be included in this study. The primary objective is to compare the efficacy of prophylactic surgery (radical colorectal resection, omentectomy, appendectomy, round ligament of the liver resection and bilateral adnexectomy) plus HIPEC CO2 with mitomycin and standard surgery in terms of local recurrence-free survival. The main secondary endpoints are disease-free survival (DFS), overall survival (OS) and safety. The primary endpoint will be described with a cumulative incidence function and will be analysed with Grey test to take account of the competing risks. DFS and OS will be described with the Kaplan-Meier method. ETHICS AND DISSEMINATION This trial has been evaluated by the Italian Medicines Agency, local ethics committees and will be submitted to the Ministry of Health to notify the start of the trial according to the regulation of trials on devices with CE mark/certification.The results will be submitted for presentation at academic meetings and for publication in a peer-reviewed journal, whatever the findings. TRIAL REGISTRATION NUMBER NCT03914820.
Collapse
Affiliation(s)
- Fabio Pacelli
- Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Chiara Gerardi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Eliana Rulli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Carlo Abatini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Stefano Rotolo
- Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche (Di.Chir.On.S.), Università degli Studi di Palermo, Palermo, Italy
| | - Silvio Garattini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | | | - Valter Torri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Fabio Galli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Erica Rulli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Andrea Di Giorgio
- Chirurgia del Peritoneo e del Retroperitoneo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| |
Collapse
|
24
|
Delhorme JB, Sauvinet G, Séverac F, Diab S, Liu D, Rohr S, Romain B, Brigand C. Peritoneal Metastases of Colorectal Origin Treated with Complete Cytoreduction and Hyperthermic Intraperitoneal Chemotherapy: The Efficiency of Mitomycin C. Ann Surg Oncol 2022; 29:7568-7576. [PMID: 35882692 DOI: 10.1245/s10434-022-12221-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/25/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Survival of patients affected by colorectal cancer peritoneal metastases (CRC-PM) can be improved with combined complete cytoreductive surgery (CCRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). Two chemotherapeutic agents are mainly used: mitomycin C (MMC) and oxaliplatin. A recent prospective randomized clinical trial showed that oxaliplatin-based HIPEC does not improve survival compared with CCRS alone. The purpose of our study was to compare the survival effectiveness of MMC versus oxaliplatin-based HIPEC using a homogeneous surgical technique and drug protocol. METHODS This retrospective monocentric study included all patients prospectively registered for having undergone CCRS and HIPEC using MMC or oxaliplatin for CRC-PM in Strasbourg University Hospital, France, from December 2004 until December 2019. MMC-based HIPEC and oxaliplatin-based HIPEC groups were compared with an inverse probability of treatment weighting. RESULTS A total of 137 patients were included. Groups were comparable for all baseline characteristics except for peritoneal carcinomatosis index. In the weighted multivariate analysis, disease-free survival (DFS) and peritoneal disease-free survival (PDFS) were significantly higher in the MMC-based HIPEC group compared with the oxaliplatin-based HIPEC group with a hazard ratio of 0.74 (CI 95% 0.56-0.98), p = 0.035 and 0.59 (CI 95% 0.40-0.98), p = 0.0084, respectively. There was no difference in overall survival or postoperative morbidity between groups. CONCLUSIONS These results favor a superiority of MMC for DFS and PDFS in comparison with oxaliplatin in HIPEC after CCRS in treatment with curative intent for CRC-PM.
Collapse
Affiliation(s)
- Jean-Baptiste Delhorme
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France. .,INSERM UMR_S1113, Université de Strasbourg, FMTS, Strasbourg, France.
| | - Guillaume Sauvinet
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France
| | - François Séverac
- Department of Public Health, Strasbourg University Hospital, Strasbourg, France
| | - Samer Diab
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France
| | - David Liu
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France
| | - Serge Rohr
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,INSERM UMR_S1113, Université de Strasbourg, FMTS, Strasbourg, France
| | - Benoît Romain
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,INSERM UMR_S1113, Université de Strasbourg, FMTS, Strasbourg, France
| | - Cécile Brigand
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,INSERM UMR_S1113, Université de Strasbourg, FMTS, Strasbourg, France
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Laoukili J, Constantinides A, Wassenaar ECE, Elias SG, Raats DAE, van Schelven SJ, van Wettum J, Volckmann R, Koster J, Huitema ADR, Nienhuijs SW, de Hingh IHJT, Wiezer RJ, van Grevenstein HMU, Rinkes IHMB, Boerma D, Kranenburg O. Peritoneal metastases from colorectal cancer belong to Consensus Molecular Subtype 4 and are sensitised to oxaliplatin by inhibiting reducing capacity. Br J Cancer 2022; 126:1824-1833. [PMID: 35194192 PMCID: PMC9174226 DOI: 10.1038/s41416-022-01742-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/19/2022] [Accepted: 02/03/2022] [Indexed: 01/13/2023] Open
Abstract
Background Peritoneal metastases (PM) in colorectal cancer (CRC) are associated with therapy resistance and poor survival. Oxaliplatin monotherapy is widely applied in the intraperitoneal treatment of PM, but fails to yield clinical benefit. We aimed to identify the mechanism(s) underlying PM resistance to oxaliplatin and to develop strategies overcoming such resistance. Experimental design We generated a biobank consisting of 35 primary tumour regions and 59 paired PM from 12 patients. All samples were analysed by RNA sequencing. We also generated a series of PM-derived organoid (PMDO) cultures and used these to design and test strategies to overcome resistance to oxaliplatin. Results PM displayed various hallmarks of aggressive CRC biology. The vast majority of PM and paired primary tumours belonged to the Consensus Molecular Subtype 4 (CMS4). PMDO cultures were resistant to oxaliplatin and expressed high levels of glutamate-cysteine ligase (GCLC) causing detoxification of oxaliplatin through glutathione synthesis. Genetic or pharmacological targeting of GCLC sensitised PMDOs to a 1-h exposure to oxaliplatin, through increased platinum-DNA adduct formation. Conclusions These results link oxaliplatin resistance of colorectal PM to their CMS4 status and high reducing capacity. Inhibiting the reducing capacity of PM may be an effective strategy to overcome PM resistance to oxaliplatin. ![]()
Collapse
Affiliation(s)
- Jamila Laoukili
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Emma C E Wassenaar
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Danielle A E Raats
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.,Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, the Netherlands
| | - Susanne J van Schelven
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jonathan van Wettum
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Richard Volckmann
- Department of Oncogenomics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jan Koster
- Department of Oncogenomics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Centre, Utrecht, the Netherlands.,Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Simon W Nienhuijs
- Department of Surgery, Catharina Hospital, Eindhoven, 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
| | - René J Wiezer
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | | | - Inne H M Borel Rinkes
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands.
| | - Onno Kranenburg
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands. .,Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
28
|
Lundbech M, Krag AE, Iversen LH, Hvas AM. Postoperative bleeding and venous thromboembolism in colorectal cancer patients undergoing cytoreductive surgery with hyperthermic intraperitoneal chemotherapy: a systematic review and meta-analysis. Int J Colorectal Dis 2022; 37:17-33. [PMID: 34626208 DOI: 10.1007/s00384-021-04021-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE Cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) has improved survival for selected patients with peritoneal metastases from colorectal cancer. Previous studies report conflicting rates of postoperative bleeding and venous thromboembolism (VTE) after CRS + HIPEC. The aim of the present study was to systematically review the literature and to estimate the overall 30-day incidence of postoperative bleeding and the overall 90-day incidence of VTE after CRS + HIPEC. METHODS Studies were identified in PubMed, Embase, and Web of Science on 29 April 2021. Data were extracted for a qualitative synthesis and to estimate an overall mean incidence in the meta-analysis. RESULTS Fourteen studies with a total of 3268 patients were included in the systematic review. Postoperative bleeding incidence rates within 30 days ranged from 1.7 to 8.3% with an overall 30-day postoperative bleeding incidence with [95% CI] at 4.2 [2.6;6.2]%. VTE incidence rates within 90 days ranged from 0.2 to 13.6% with an overall 90-day VTE incidence with [95% CI] at 2.7 [1;5.2]% after CRS + HIPEC. CONCLUSION This systematic review and meta-analysis indicate a low risk for postoperative bleeding within 30 days and VTE within 90 days after CRS + HIPEC for peritoneal metastases from colorectal cancer.
Collapse
Affiliation(s)
- Mikkel Lundbech
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Andreas Engel Krag
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.,Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Lene Hjerrild Iversen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark. .,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
29
|
Abstract
Peritoneal surface malignancies comprise a heterogeneous group of primary tumours, including peritoneal mesothelioma, and peritoneal metastases of other tumours, including ovarian, gastric, colorectal, appendicular or pancreatic cancers. The pathophysiology of peritoneal malignancy is complex and not fully understood. The two main hypotheses are the transformation of mesothelial cells (peritoneal primary tumour) and shedding of cells from a primary tumour with implantation of cells in the peritoneal cavity (peritoneal metastasis). Diagnosis is challenging and often requires modern imaging and interventional techniques, including surgical exploration. In the past decade, new treatments and multimodal strategies helped to improve patient survival and quality of life and the premise that peritoneal malignancies are fatal diseases has been dismissed as management strategies, including complete cytoreductive surgery embedded in perioperative systemic chemotherapy, can provide cure in selected patients. Furthermore, intraperitoneal chemotherapy has become an important part of combination treatments. Improving locoregional treatment delivery to enhance penetration to tumour nodules and reduce systemic uptake is one of the most active research areas. The current main challenges involve not only offering the best treatment option and developing intraperitoneal therapies that are equivalent to current systemic therapies but also defining the optimal treatment sequence according to primary tumour, disease extent and patient preferences. New imaging modalities, less invasive surgery, nanomedicines and targeted therapies are the basis for a new era of intraperitoneal therapy and are beginning to show encouraging outcomes.
Collapse
|
30
|
Mangieri CW, Moaven O, Valenzuela CD, Erali RA, Votanopoulos KI, Shen P, Levine EA. Utility of hyperthermic intraperitoneal chemotherapy in cases of incomplete cytoreductive surgery. J Surg Oncol 2021; 125:703-711. [PMID: 34841542 DOI: 10.1002/jso.26759] [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] [Received: 09/17/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Hyperthermic intraperitoneal chemotherapy (HIPEC) during cytoreductive surgery (CRS) is typically reserved for a complete or optimal cytoreduction. There is the potential for therapeutic effect of HIPEC with an incomplete cytoreduction, particularly for near optimal cytoreductions. METHODS Retrospective review of incomplete cytoreductions (R2b, R2c) for appendiceal and colorectal primaries. Primary endpoints were overall survival (OS) and progression-free survival (PFS). Subgroup analysis for primary etiology and specific cytoreductive score. RESULTS A total of 121 cases of incomplete CRS, 74 CRS alone, and 47 CRS-HIPEC. For the entire study group there was a survival benefit with HIPEC. OS and PFS were 2.3 versus 1.4 (p = 0.001) and 1.6 versus 0.7 (p < 0.0001) respectively for cases with and without HIPEC. Subgroup analysis of appendiceal neoplasms, 43 CRS-HIPEC and 50 CRS alone, found HIPEC benefit persisted; OS and PFS were 2.4 versus 1.5 (p = 0.016) and 1.7 versus 0.8 (p < 0.0001), respectively for cases with and without HIPEC. Benefit most pronounced in low-grade cases with doubling of the OS and PFS (p = 0.004). With colorectal primary cases, 10 CRS-HIPEC and 18 CRS alone, no difference in OS and PFS. When stratifying out by cytoreduction scores, R2b and R2c, HIPEC only provided a benefit for R2b cases; OS and PFS for R2b cases were 2.28 versus 1.01 (p = 0.011) and 1.67 versus 0.75 (p = 0.001), respectively for cases with and without HIPEC. CONCLUSION HIPEC has utility for incomplete cytoreductions with appendiceal neoplasms, greatest effect with low-grade appendiceal neoplasms. HIPEC is only beneficial for near optimal cytoreductions (R2b).
Collapse
Affiliation(s)
- Christopher W Mangieri
- Division of Surgical Oncology, Surgical Oncology Section, Wake Forest Baptist Health Medical Center, Winston-Salem, North Carolina, USA
| | - Omeed Moaven
- Department of Surgery, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Cristian D Valenzuela
- Division of Surgical Oncology, Surgical Oncology Section, Wake Forest Baptist Health Medical Center, Winston-Salem, North Carolina, USA
| | - Richard A Erali
- Division of Surgical Oncology, Surgical Oncology Section, Wake Forest Baptist Health Medical Center, Winston-Salem, North Carolina, USA
| | - Konstantinos I Votanopoulos
- Division of Surgical Oncology, Surgical Oncology Section, Wake Forest Baptist Health Medical Center, Winston-Salem, North Carolina, USA
| | - Perry Shen
- Division of Surgical Oncology, Surgical Oncology Section, Wake Forest Baptist Health Medical Center, Winston-Salem, North Carolina, USA
| | - Edward A Levine
- Division of Surgical Oncology, Surgical Oncology Section, Wake Forest Baptist Health Medical Center, Winston-Salem, North Carolina, USA
| |
Collapse
|
31
|
Sun T, Zhang G, Ning T, Chen Q, Chu Y, Luo Y, You H, Su B, Li C, Guo Q, Jiang C. A Versatile Theranostic Platform for Colorectal Cancer Peritoneal Metastases: Real-Time Tumor-Tracking and Photothermal-Enhanced Chemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102256. [PMID: 34398516 PMCID: PMC8529449 DOI: 10.1002/advs.202102256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/30/2021] [Indexed: 06/13/2023]
Abstract
A versatile tumor-targeting stimuli-responsive theranostic platform for peritoneal metastases of colorectal cancer is proposed in this work for tumor tracking and photothermal-enhanced chemotherapy. A quenched photosensitizer ("off" state) is developed and escorted into a tumor-targeting oxaliplatin-embedded micelle. Once reaching the tumor cell, the micelle is clasped to release free oxaliplatin, as well as the "off" photosensitizer, which is further activated ("turned-on") in the tumor reducing microenvironment to provide optical imaging and photothermal effect. The combined results from hyperthermia-enhanced chemotherapy, deep penetration, perfused O2 , and the leveraged GSH-ROS imbalance in tumor cells are achieved for improved antitumor efficacy and reduced systematic toxicity.
Collapse
Affiliation(s)
- Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Guangping Zhang
- Shandong Province Key Laboratory of Medical Physics and Image Processing TechnologySchool of Physics and Electronics and Institute of Materials and Clean EnergyShandong Normal University1 University RoadJinan250358P. R. China
| | - Tingting Ning
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Yongchao Chu
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Yifan Luo
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Haoyu You
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Boyu Su
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Chao Li
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Qin Guo
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education)Minhang HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceDepartment of PharmaceuticsSchool of PharmacyFudan University826 Zhangheng RoadShanghai201203China
| |
Collapse
|
32
|
Lee YS, Lee WS, Kim CW, Lee SJ, Yang H, Kong SJ, Ning J, Yang KM, Kang B, Kim WR, Chon HJ, Kim C. Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer. J Immunother Cancer 2021; 8:jitc-2020-000857. [PMID: 33199510 PMCID: PMC7670945 DOI: 10.1136/jitc-2020-000857] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/14/2022] Open
Abstract
Background Peritoneal carcinomatosis (PC) is a common and devastating manifestation of colon cancer and refractory to conventional anticancer therapeutics. During the peritoneal dissemination of colon cancer, peritoneal immunity is nullified by various mechanisms of immune evasion. Here, we employed the armed oncolytic vaccinia virus mJX-594 (JX) to rejuvenate the peritoneal antitumor immune responses in the treatment of PC. Methods PC model of MC38 colon cancer was generated and intraperitoneally treated with JX and/or anti-programmed cell death protein 1 (PD-1) antibody. The peritoneal tumor burden, vascular leakage, and malignant ascites formation were then assessed. Tumors and peritoneal lavage cells were analyzed by flow cytometry, multiplex tissue imaging, and a NanoString assay. Results JX treatment effectively suppressed peritoneal cancer progression and malignant ascites formation. It also restored the peritoneal anticancer immunity by activating peritoneal dendritic cells (DCs) and CD8+ T cells. Moreover, JX selectively infected and killed peritoneal colon cancer cells and promoted the intratumoral infiltration of DCs and CD8+ T cells into peritoneal tumor nodules. JX reinvigorates anticancer immunity by reprogramming immune-related transcriptional signatures within the tumor microenvironment. Notably, JX cooperates with immune checkpoint inhibitors (ICIs), anti-programmed death-1, anti-programmed death-ligand 1, and anti-lymphocyte-activation gene-3 to elicit a stronger anticancer immunity that eliminates peritoneal metastases and malignant ascites of colon cancer compared with JX or ICI alone. Conclusions Intraperitoneal immunotherapy with JX restores peritoneal anticancer immunity and potentiates immune checkpoint blockade to suppress PC and malignant ascites in colon cancer.
Collapse
Affiliation(s)
- Yu Seong Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea (the Republic of)
| | - Won Suk Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Chang Woo Kim
- Kyung Hee University Gangdong Hospital, Gangdong-gu, Korea (the Republic of)
| | - Seung Joon Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea (the Republic of)
| | - Hannah Yang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - So Jung Kong
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - John Ning
- SillaJen Biotherapeutics, San Francisco, California, USA
| | | | - Beodeul Kang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Woo Ram Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea (the Republic of)
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
McCarthy B, Singh R, Levi-Polyachenko N. Oxaliplatin-resistant colorectal cancer models for nanoparticle hyperthermia. Int J Hyperthermia 2021; 38:152-164. [PMID: 33576281 DOI: 10.1080/02656736.2021.1876253] [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 Metastatic colorectal cancer (CRC) is complicated by chemotherapy-resistant cell populations. Oxaliplatin is used in heated intraperitoneal hyperthermic chemoperfusion (HIPEC) for treatment of disseminated CRC. Photothermal nanoparticles can provide focal heating to improve the response of CRC cells to oxaliplatin, by confining heating near individual cells. Reduction in cellular luciferase signal may allow single-cell-resolution recording of thermal dosimetry. METHODS Oxaliplatin resistant (OxR) variants of luciferase-expressing CT26.WT-Fluc-Neo CRC cells were developed and their sensitivity to hyperthermia was evaluated. Polymer-based photothermal nanoparticles were developed, characterized and used to explore their potential for imparting a thermal dose to improve cell response to oxaliplatin. A correlation of thermal dose to intracellular luciferase activity was established using quantitative luminescence monitoring and microscopy. RESULTS Luciferase-based monitoring of thermal dose within CT26 cell lines was validated within the ranges of 0.04-8.33 CEM43 for parental cells and 0.05-9.74 CEM43 for OxR CT26 cells. This was further confirmed using nanoparticle-induced hyperthermia, where the single-cell resolution of the thermal dose can be achieved. The nanoparticles enhance cell killing of resistant cells when combined with oxaliplatin and stimulated to generate heat. CONCLUSION Nanoparticle-based hyperthermia is effective for augmenting chemotherapy and can be coupled with reductions in CT26 luciferase expression to monitor thermal dose at single-cell resolution. The development of OxR CT26.WT-Fluc-Neo CRC cells sets the stage for pre-clinical evaluations to measure nanoparticle-induced hyperthermia to augment chemotherapy (Nano-HIPEC) in a chemotherapy-resistant model of disseminated CRC.
Collapse
Affiliation(s)
- Bryce McCarthy
- Department of Plastic and Reconstructive Surgery Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nicole Levi-Polyachenko
- Department of Plastic and Reconstructive Surgery Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
36
|
Kok HP, Cressman ENK, Ceelen W, Brace CL, Ivkov R, Grüll H, Ter Haar G, Wust P, Crezee J. Heating technology for malignant tumors: a review. Int J Hyperthermia 2021; 37:711-741. [PMID: 32579419 DOI: 10.1080/02656736.2020.1779357] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The therapeutic application of heat is very effective in cancer treatment. Both hyperthermia, i.e., heating to 39-45 °C to induce sensitization to radiotherapy and chemotherapy, and thermal ablation, where temperatures beyond 50 °C destroy tumor cells directly are frequently applied in the clinic. Achievement of an effective treatment requires high quality heating equipment, precise thermal dosimetry, and adequate quality assurance. Several types of devices, antennas and heating or power delivery systems have been proposed and developed in recent decades. These vary considerably in technique, heating depth, ability to focus, and in the size of the heating focus. Clinically used heating techniques involve electromagnetic and ultrasonic heating, hyperthermic perfusion and conductive heating. Depending on clinical objectives and available technology, thermal therapies can be subdivided into three broad categories: local, locoregional, or whole body heating. Clinically used local heating techniques include interstitial hyperthermia and ablation, high intensity focused ultrasound (HIFU), scanned focused ultrasound (SFUS), electroporation, nanoparticle heating, intraluminal heating and superficial heating. Locoregional heating techniques include phased array systems, capacitive systems and isolated perfusion. Whole body techniques focus on prevention of heat loss supplemented with energy deposition in the body, e.g., by infrared radiation. This review presents an overview of clinical hyperthermia and ablation devices used for local, locoregional, and whole body therapy. Proven and experimental clinical applications of thermal ablation and hyperthermia are listed. Methods for temperature measurement and the role of treatment planning to control treatments are discussed briefly, as well as future perspectives for heating technology for the treatment of tumors.
Collapse
Affiliation(s)
- H Petra Kok
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik N K Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wim Ceelen
- Department of GI Surgery, Ghent University Hospital, Ghent, Belgium
| | - Christopher L Brace
- Department of Radiology and Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Holger Grüll
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Gail Ter Haar
- Department of Physics, The Institute of Cancer Research, London, UK
| | - Peter Wust
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Ceelen W, Demuytere J, de Hingh I. Hyperthermic Intraperitoneal Chemotherapy: A Critical Review. Cancers (Basel) 2021; 13:cancers13133114. [PMID: 34206563 PMCID: PMC8268659 DOI: 10.3390/cancers13133114] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Patients with cancer of the digestive system or ovarian cancer are at risk of developing peritoneal metastases (PM). In some patients with PM, surgery followed by intraperitoneal (IP) chemotherapy has emerged as a valid treatment option. The addition of hyperthermia is thought to further enhance the efficacy of IP chemotherapy. However, the results of recent clinical trials in large bowel cancer have put into question the use of hyperthermic intraperitoneal chemotherapy (HIPEC). Here, we review the rationale and current results of HIPEC for PM and propose a roadmap to further progress. Abstract With increasing awareness amongst physicians and improved radiological imaging techniques, the peritoneal cavity is increasingly recognized as an important metastatic site in various malignancies. Prognosis of these patients is usually poor as traditional treatment including surgical resection or systemic treatment is relatively ineffective. Intraperitoneal delivery of chemotherapeutic agents is thought to be an attractive alternative as this results in high tumor tissue concentrations with limited systemic exposure. The addition of hyperthermia aims to potentiate the anti-tumor effects of chemotherapy, resulting in the concept of heated intraperitoneal chemotherapy (HIPEC) for the treatment of peritoneal metastases as it was developed about 3 decades ago. With increasing experience, HIPEC has become a safe and accepted treatment offered in many centers around the world. However, standardization of the technique has been poor and results from clinical trials have been equivocal. As a result, the true value of HIPEC in the treatment of peritoneal metastases remains a matter of debate. The current review aims to provide a critical overview of the theoretical concept and preclinical and clinical study results, to outline areas of persisting uncertainty, and to propose a framework to better define the role of HIPEC in the treatment of peritoneal malignancies.
Collapse
Affiliation(s)
- Wim Ceelen
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9332-6251
| | - Jesse Demuytere
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Ignace de Hingh
- Department of Surgery, Catharina Cancer Institute, PO Box 1350, 5602 ZA Eindhoven, The Netherlands;
- GROW—School for Oncology and Developmental Biology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
38
|
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
|
39
|
Yurttas C, Horvath P, Fischer I, Meisner C, Nadalin S, Königsrainer I, Königsrainer A, Beckert S, Löffler MW. A Prospective, Phase I/II, Open-Label Pilot Trial to Assess the Safety of Hyperthermic Intraperitoneal Chemotherapy After Oncological Resection of Pancreatic Adenocarcinoma. Ann Surg Oncol 2021; 28:9086-9095. [PMID: 34131821 PMCID: PMC8205203 DOI: 10.1245/s10434-021-10187-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a common fatal disease with unfavorable prognosis, even after oncological resection. To improve survival, adding hyperthermic intraperitoneal chemotherapy (HIPEC) has been suggested. Whether HIPEC entails disproportional short-term mortality is unknown and a prospectively determined adverse events profile is lacking. Since both pancreatic resection and HIPEC may relevantly influence morbidity and mortality, this uncontrolled single-arm, open-label, phase I/II pilot trial was designed to assess the 30-day mortality rate, treatment feasibility, and adverse events connected with HIPEC after oncological pancreatic surgery. METHODS This trial recruited patients scheduled for PDAC resection. A sample size of 16 patients receiving study interventions was estimated to establish a predefined margin of treatment-associated short-term mortality with a power of > 80%. Patients achieving complete macroscopic resection received HIPEC with gemcitabine administered at 1000 mg/m2 body surface area heated to 42 °C for 1 hour. RESULTS Within 30 days after intervention, no patient died or experienced any adverse events higher than grade 3 that were related to HIPEC. Furthermore, treatment-related adverse events were prospectively documented and categorized as expected or unexpected. This trial supports that the actual mortality rate after PDAC resection and HIPEC is below 10%. HIPEC treatment proved feasible in 89% of patients allocated to intervention. Pancreatic fistulas, as key complications after pancreas surgery, occurred in 3/13 patients under risk. CONCLUSION Combined pancreas resection and gemcitabine HIPEC proved feasible and safe, with acceptable morbidity and mortality. Based on these results, further clinical evaluation can be justified. REGISTRATION NUMBER NCT02863471 ( http://www.clinicaltrials.gov ).
Collapse
Affiliation(s)
- Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Philipp Horvath
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Imma Fischer
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Meisner
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Ingmar Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany.,Department of General, Visceral and Thoracic Surgery, Landeskrankenhaus Feldkirch, Feldkirch, Austria
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Stefan Beckert
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany.,Department of General and Visceral Surgery, Schwarzwald-Baar Klinikum Villingen-Schwenningen, Villingen-Schwenningen, Germany
| | - Markus W Löffler
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany. .,Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany. .,Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany. .,Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
40
|
Tsai TY, You JF, Hsu YJ, Jhuang JR, Chern YJ, Hung HY, Yeh CY, Hsieh PS, Chiang SF, Lai CC, Chiang JM, Tang R, Tsai WS. A Prediction Model for Metachronous Peritoneal Carcinomatosis in Patients with Stage T4 Colon Cancer after Curative Resection. Cancers (Basel) 2021; 13:2808. [PMID: 34200032 PMCID: PMC8200190 DOI: 10.3390/cancers13112808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 01/21/2023] Open
Abstract
(1) Background: The aim of this study was to develop a prediction model for assessing individual mPC risk in patients with pT4 colon cancer. Methods: A total of 2003 patients with pT4 colon cancer undergoing R0 resection were categorized into the training or testing set. Based on the training set, 2044 Cox prediction models were developed. Next, models with the maximal C-index and minimal prediction error were selected. The final model was then validated based on the testing set using a time-dependent area under the curve and Brier score, and a scoring system was developed. Patients were stratified into the high- or low-risk group by their risk score, with the cut-off points determined by a classification and regression tree (CART). (2) Results: The five candidate predictors were tumor location, preoperative carcinoembryonic antigen value, histologic type, T stage and nodal stage. Based on the CART, patients were categorized into the low-risk or high-risk groups. The model has high predictive accuracy (prediction error ≤5%) and good discrimination ability (area under the curve >0.7). (3) Conclusions: The prediction model quantifies individual risk and is feasible for selecting patients with pT4 colon cancer who are at high risk of developing mPC.
Collapse
Affiliation(s)
- Tzong-Yun Tsai
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Jeng-Fu You
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Yu-Jen Hsu
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Jing-Rong Jhuang
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei City 10055, Taiwan;
| | - Yih-Jong Chern
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Hsin-Yuan Hung
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Chien-Yuh Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Pao-Shiu Hsieh
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Sum-Fu Chiang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Cheng-Chou Lai
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Jy-Ming Chiang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Reiping Tang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| | - Wen-Sy Tsai
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (T.-Y.T.); (J.-F.Y.); (Y.-J.H.); (Y.-J.C.); (H.-Y.H.); (C.-Y.Y.); (P.-S.H.); (S.-F.C.); (C.-C.L.); (J.-M.C.); (R.T.)
- College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan
| |
Collapse
|
41
|
Rovers KP, Bakkers C, Nienhuijs SW, Burger JWA, Creemers GJM, Thijs AMJ, Brandt-Kerkhof ARM, Madsen EVE, van Meerten E, Tuynman JB, Kusters M, Versteeg KS, Aalbers AGJ, Kok NFM, Buffart TE, Wiezer MJ, Boerma D, Los M, de Reuver PR, Bremers AJA, Verheul HMW, Kruijff S, de Groot DJA, Witkamp AJ, van Grevenstein WMU, Koopman M, Nederend J, Lahaye MJ, Kranenburg O, Fijneman RJA, van 't Erve I, Snaebjornsson P, Hemmer PHJ, Dijkgraaf MGW, Punt CJA, Tanis PJ, de Hingh IHJT. Perioperative Systemic Therapy vs Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy Alone for Resectable Colorectal Peritoneal Metastases: A Phase 2 Randomized Clinical Trial. JAMA Surg 2021; 156:710-720. [PMID: 34009291 DOI: 10.1001/jamasurg.2021.1642] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Importance To date, no randomized clinical trials have investigated perioperative systemic therapy relative to cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) alone for resectable colorectal peritoneal metastases (CPM). Objective To assess the feasibility and safety of perioperative systemic therapy in patients with resectable CPM and the response of CPM to neoadjuvant treatment. Design, Setting, and Participants An open-label, parallel-group phase 2 randomized clinical trial in all 9 Dutch tertiary centers for the surgical treatment of CPM enrolled participants between June 15, 2017, and January 9, 2019. Participants were patients with pathologically proven isolated resectable CPM who did not receive systemic therapy within 6 months before enrollment. Interventions Randomization to perioperative systemic therapy or CRS-HIPEC alone. Perioperative systemic therapy comprised either four 3-week neoadjuvant and adjuvant cycles of CAPOX (capecitabine and oxaliplatin), six 2-week neoadjuvant and adjuvant cycles of FOLFOX (fluorouracil, leucovorin, and oxaliplatin), or six 2-week neoadjuvant cycles of FOLFIRI (fluorouracil, leucovorin, and irinotecan) and either four 3-week adjuvant cycles of capecitabine or six 2-week adjuvant cycles of fluorouracil with leucovorin. Bevacizumab was added to the first 3 (CAPOX) or 4 (FOLFOX/FOLFIRI) neoadjuvant cycles. Main Outcomes and Measures Proportions of macroscopic complete CRS-HIPEC and Clavien-Dindo grade 3 or higher postoperative morbidity. Key secondary outcomes were centrally assessed rates of objective radiologic and major pathologic response of CPM to neoadjuvant treatment. Analyses were done modified intention-to-treat in patients starting neoadjuvant treatment (experimental arm) or undergoing upfront surgery (control arm). Results In 79 patients included in the analysis (43 [54%] men; mean [SD] age, 62 [10] years), experimental (n = 37) and control (n = 42) arms did not differ significantly regarding the proportions of macroscopic complete CRS-HIPEC (33 of 37 [89%] vs 36 of 42 [86%] patients; risk ratio, 1.04; 95% CI, 0.88-1.23; P = .74) and Clavien-Dindo grade 3 or higher postoperative morbidity (8 of 37 [22%] vs 14 of 42 [33%] patients; risk ratio, 0.65; 95% CI, 0.31-1.37; P = .25). No treatment-related deaths occurred. Objective radiologic and major pathologic response rates of CPM to neoadjuvant treatment were 28% (9 of 32 evaluable patients) and 38% (13 of 34 evaluable patients), respectively. Conclusions and Relevance In this randomized phase 2 trial in patients diagnosed with resectable CPM, perioperative systemic therapy seemed feasible, safe, and able to induce response of CPM, justifying a phase 3 trial. Trial Registration ClinicalTrials.gov Identifier: NCT02758951.
Collapse
Affiliation(s)
- Koen P Rovers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Checca Bakkers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Simon W Nienhuijs
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Jacobus W A Burger
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Geert-Jan M Creemers
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Anna M J Thijs
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, the Netherlands
| | | | - Eva V E Madsen
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jurriaan B Tuynman
- Department of Surgery, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands
| | - Miranda Kusters
- Department of Surgery, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands
| | - Kathelijn S Versteeg
- Department of Medical Oncology, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands
| | - Arend G J Aalbers
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Niels F M Kok
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tineke E Buffart
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marinus J Wiezer
- Department of Surgery, Sint Antonius Hospital, Nieuwegein, the Netherlands
| | - Djamila Boerma
- Department of Surgery, Sint Antonius Hospital, Nieuwegein, the Netherlands
| | - Maartje Los
- Department of Medical Oncology, Sint Antonius Hospital, Nieuwegein, the Netherlands
| | - Philip R de Reuver
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas J A Bremers
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Schelto Kruijff
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Arjen J Witkamp
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, 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
| | - Onno Kranenburg
- Cancer Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Remond J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Iris van 't Erve
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Patrick H J Hemmer
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Marcel G W Dijkgraaf
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Cornelis J A Punt
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Pieter J Tanis
- Department of Surgery, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, 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
|
42
|
Twelve-Year Single Center Experience Shows Safe Implementation of Developed Peritoneal Surface Malignancy Treatment Protocols for Gastrointestinal and Gynecological Primary Tumors. Cancers (Basel) 2021; 13:cancers13102471. [PMID: 34069475 PMCID: PMC8159136 DOI: 10.3390/cancers13102471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The treatment of peritoneal surface malignancies (PSM) has dramatically evolved during the past two decades. Indications, treatment protocols, surgical techniques and the application of HIPEC in the prophylactic setting were evaluated in the surgical community. Nevertheless, the current results of the PRODIGE-7 trial disfavored the application of HIPEC for PSM of colorectal cancer and raised uncertainty among surgeons. On the other hand, cytoreductive surgery and HIPEC represent state-of-the-art therapy for peritoneal mesothelioma (except the sarcomatoid-subtype) and pseudomyxoma peritonei. Comparing the literature is cumbersome due to the variety of HIPEC protocols and differences in indication settings. This article aims to provide an insight into the impact of different HIPEC protocols, different indication settings and the implementation of pre-HIPEC laparoscopy on patients’ morbidity rates and outcomes and serves as guidance for surgeons dealing with these patients in order to guarantee high-quality treatment. Abstract (1) Background: Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy provide survival benefits to selected patients. We aimed to report our experience and the evolution of our peritoneal surface malignancy program. (2) Methods: From June 2005 to June 2017, 399 patients who underwent cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy at the Tübingen University Hospital were analyzed from a prospectively collected database. (3) Results: Peritoneal metastasis from colorectal cancer was the leading indication (group 1: 28%; group 2: 32%). The median PCI was 15.5 (range, 1–39) in group 1 and 11 (range, 1–39) in group 2 (p = 0.002). Regarding the completeness of cytoreduction (CC), a score of 0 was achieved in 63% vs. 69% for group 1 and 2, respectively (p = 0.010). Median overall survival rates for patients in group 1 and 2 for colon cancer, ovarian cancer, gastric cancer and appendix cancer were 34 and 25 months; 45 months and not reached; 30 and 16 months; 39 months and not reached, respectively. The occurrence of grade-III and -IV complications slightly differed between groups (14.5% vs. 15.6%). No 30-day mortality occurred. (4) Conclusions: Specialized centers are able to provide low-morbidity cytoreductive surgery and hyperthermic intraperitoneal chemotherapy without mortality. Strict patient selection during the time period significantly improved CC scores.
Collapse
|
43
|
Carboni F, Valle M. Limitations of the PRODIGE 7 trial. Lancet Oncol 2021; 22:e176. [PMID: 33932374 DOI: 10.1016/s1470-2045(21)00088-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Fabio Carboni
- Peritoneal Tumours Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Mario Valle
- Peritoneal Tumours Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| |
Collapse
|
44
|
Cytoreductive surgery and mitomycin C hyperthermic intraperitoneal chemotherapy with CO 2 recirculation (HIPEC-CO 2) for colorectal cancer peritoneal metastases: analysis of short-term outcomes. Updates Surg 2021; 73:1443-1448. [PMID: 33782856 DOI: 10.1007/s13304-021-01034-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/16/2021] [Indexed: 12/27/2022]
Abstract
Peritoneal dissemination from colorectal cancer (CRC) has long been associated with unfavorable prognosis. However, in the last decades, the combination of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) was able to obtain up to 30% 5-year survival rate in selected centers. Despite the wide diffusion of CRS and HIPEC, until now, there are no clear recommendations on the drug of choice for HIPEC nor its technique, and safety and efficacy data of HIPEC regimens and techniques are lacking. We performed a retrospective analysis of a prospectively maintained database of 26 CRS and mitomycin C HIPEC with CO2 recirculation (HIPEC-CO2) for CRC peritoneal metastasis (PM) performed at our center. The main endpoints were morbidity, mortality, the temperature of perfusate during HIPEC and metabolic changes throughout the procedure. Morbidity was assessed by analysis of postoperative adverse events according to the Common Terminology Criteria for Adverse Events (CTCAE version 4.0). Continuous variables of Arterial Blood Gas (ABG) analysis at three time-points were compared by the Student t test. There were no postoperative deaths. The overall grade 3-4 CTCAE complications rate at 30 days was 38.4%, with ten severe adverse events occurring to six (23.0%) patients. The temperature within HIPEC perfusion maintained between 41 and 42 °C in all cases and we experienced no HIPEC-related intraoperative complications. We observed a significant difference between all baseline and pre-HIPEC ABG parameters evaluated but no statistically significant differences between pre- and post-HIPEC ABG outcomes. This study shows that mitomycin C HIPEC-CO2 is feasible and has a safety profile comparable to that of other HIPEC techniques reported in the literature. Further research is needed to validate prospectively the safety and efficacy of this technique.
Collapse
|
45
|
Kusamura S, Barretta F, Yonemura Y, Sugarbaker PH, Moran BJ, Levine EA, Goere D, Baratti D, Nizri E, Morris DL, Glehen O, Sardi A, Barrios P, Quénet F, Villeneuve L, Gómez-Portilla A, de Hingh I, Ceelen W, Pelz JOW, Piso P, González-Moreno S, Van Der Speeten K, Deraco M. The Role of Hyperthermic Intraperitoneal Chemotherapy in Pseudomyxoma Peritonei After Cytoreductive Surgery. JAMA Surg 2021; 156:e206363. [PMID: 33502455 DOI: 10.1001/jamasurg.2020.6363] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Importance Studies on the prognostic role of hyperthermic intraperitoneal chemotherapy (HIPEC) in pseudomyxoma peritonei (PMP) are currently not available. Objectives To evaluate outcomes after cytoreductive surgery (CRS) and HIPEC compared with CRS alone in patients with PMP. Design, Setting, and Participants This cohort study analyzed data from the Peritoneal Surface Oncology Group International (PSOGI) registry, including 1924 patients with histologically confirmed PMP due to an appendiceal mucinous neoplasm. Eligible patients were treated with CRS with or without HIPEC from February 1, 1993, to December 31, 2017, and had complete information on the main prognostic factors and intraperitoneal treatments. Inverse probability treatment weights based on the propensity score for HIPEC treatment containing the main prognostic factors were applied to all models to balance comparisons between the CRS-HIPEC vs CRS-alone groups in the entire series and in the following subsets: optimal cytoreduction, suboptimal cytoreduction, high- and low-grade histologic findings, and different HIPEC drug regimens. Data were analyzed from March 1 to June 1, 2018. Interventions HIPEC including oxaliplatin plus combined fluorouracil-leucovorin, cisplatin plus mitomycin, mitomycin, and other oxaliplatin-based regimens. Main Outcomes and Measures Overall survival, severe morbidity (determined using the National Cancer Institute Common Terminology for Adverse Events, version 3.0), return to operating room, and 30- and 90-day mortality. Differences in overall survival were compared using weighted Kaplan-Meier curves, log-rank tests, and Cox proportional hazards multivariable models. A sensitivity analysis was based on the E-value from the results of the main Cox proportional hazards model. Differences in surgical outcomes were compared using weighted multivariable logistic models. Results Of the 1924 patients included in the analysis (997 [51.8%] men; median age, 56 [interquartile range extremes (IQRE), 45-65] years), 376 were in the CRS-alone group and 1548 in the CRS-HIPEC group. Patients with CRS alone were older (median age, 60 [IQRE, 48-70] vs 54 [IQRE, 44-63] years), had less lymph node involvement (14 [3.7%] vs 119 [7.7%]), received more preoperative systemic chemotherapy (198 [52.7%] vs 529 [34.2%]), and had higher proportions of high-grade disease (179 [47.6%] vs 492 [31.8%]) and suboptimal cytoreduction residual disease (grade 3, 175 [46.5%] vs 117 [7.6%]). HIPEC was not associated with a higher risk of worse surgical outcomes except with mitomycin, with higher odds of morbidity (1.99; 95% CI, 1.25-3.19; P = .004). HIPEC was associated with a significantly better overall survival in all subsets (adjusted hazard ratios [HRs], 0.60-0.68, with 95% CIs not crossing 1.00). The weighted 5-year overall survival was 57.8% (95% CI, 50.8%-65.7%) vs 46.2% (95% CI, 40.3%-52.8%) for CRS-HIPEC and CRS alone, respectively (weighted HR, 0.65; 95% CI, 0.50-0.83; P < .001; E-value, 2.03). Such prognostic advantage was associated with oxaliplatin plus fluorouracil-leucovorin (HR, 0.42; 95% CI, 0.19-0.93; P = .03) and cisplatin plus mitomycin (HR, 0.57; 95% CI, 0.42-0.78; P = .001) schedules. Conclusions and Relevance In this cohort study, HIPEC was associated with better overall survival when performed after CRS in PMP, generally without adverse effects on surgical outcomes.
Collapse
Affiliation(s)
- Shigeki Kusamura
- Peritoneal Surface Malignancies Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori dei Tumori di Milano, Milano, Italy
| | - Francesco Barretta
- Clinical Epidemiology and Trial Organization Unit, Fondazione IRCCS Istituto Nazionale Tumori dei Tumori di Milano, Milano, Italy
| | - Yutaka Yonemura
- Nonprofit Organization to Support Peritoneal Surface Malignancy Treatment, Kishiwada, Japan
| | | | - Brendan John Moran
- Basingstoke and North Hampshire National Health Service Foundation Trust, Basingstoke, United Kingdom
| | - Edward A Levine
- Surgical Oncology Service, Wake Forest University Baptist Medical Center, Winston-Salem, North Carolina
| | - Diane Goere
- Department of Visceral and Oncologic Surgery, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Dario Baratti
- Peritoneal Surface Malignancies Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori dei Tumori di Milano, Milano, Italy
| | - Eran Nizri
- Peritoneal Surface Malignancies Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori dei Tumori di Milano, Milano, Italy.,fellow at European School of Peritoneal Surface Oncology, Milano, Italy
| | - David Lawson Morris
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, University of New South Wales, St George Hospital, Sydney, Australia
| | - Olivier Glehen
- Department of Digestive Surgery, Centre Hospitalo-Universitaire Lyon-Sud, Hospices Civils de Lyon, Lyon, France
| | - Armando Sardi
- Division of Surgery, Department of Surgical Oncology, The Institute for Cancer Care, Mercy Medical Center, Baltimore, Maryland
| | - Pedro Barrios
- Department of Oncological Surgery, Hospital Sant Joan Despí, Moises Broggi, Peritoneal Surface Malignancy Catalonian's Programme, Sant Joan Despí, Barcelona, Spain
| | - François Quénet
- Centre Régional de Lutte Contre le Cancer Val d'Aurell, Montpellier, France
| | - Laurent Villeneuve
- Réseau National de Prise en Charge des Tumeurs Rares du Péritoine, French National Registry of Rare Peritoneal Surface Malignancies, Lyon, France
| | - Alberto Gómez-Portilla
- Department of General Surgery, Hospital Universitario de Araba, Hospital Universitario Araba Sede Hospital Santiago, Santiago, Spain.,Departamento de Cirugía General, Universidad del País Vasco, Vitoria, Spain.,Programa de Carcinomatosis Peritoneal, Hospital San José, Vitoria, Spain
| | - Ignace de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Wim Ceelen
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Joerg O W Pelz
- Department of Surgery I, University of Wuerzburg, Wuerzburg, Germany
| | - Pompiliu Piso
- Department of General and Visceral Surgery, Krankenhaus Barmherzige Brüder, Regensburg, Germany
| | - Santiago González-Moreno
- Peritoneal Surface Oncology Program, Department of Surgical Oncology, MD Anderson Cancer Center, Madrid, Spain
| | | | - Marcello Deraco
- Peritoneal Surface Malignancies Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori dei Tumori di Milano, Milano, Italy
| | | |
Collapse
|
46
|
Moral ÁSD, Viejo EP, Romero IM, Pérez FP. Results of systematic second-look surgery plus hipec in perforated or pt4 colon cancer. Case series. Ann Med Surg (Lond) 2021; 62:386-390. [PMID: 33552500 PMCID: PMC7851413 DOI: 10.1016/j.amsu.2021.01.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 11/29/2022] Open
Abstract
Background Perforated or pT4 colonic tumors have a bad prognosis with a high rate of relapse, including peritoneal relapse (20–30%). Our aim is to analyze the effectiveness of Second Look surgery (SLS) + hyperthermic intraperitoneal chemotherapy (HIPEC) in these patients for early treatment of peritoneal relapse (PR) or for preventing it. Patients and methods Patients previously operated for colon cancer, either pT4 or perforated (M0), with no evidence of disease at any level after adjuvant chemotherapy, who undergo systematic SLS + HIPEC (Oxaliplatin 30 min) one year after the initial surgery. Results Since February 2014 to July 2018, we performed SLS + HIPEC in 42 patients with M0, either pT4 (n = 33) or perforated (n = 9) colon cancer. Although during SLS there were suspicious lesions in 15 cases (37.5%), they were histologically confirmed in only 4 (9.5%). Histologically confirmed peritoneal relapse (PR) rate at SLS was 6% in pT4 (2/33) and 22.2% in perforated tumors (2/9). Prophylactic HIPEC was performed in all the cases. There was no postoperative mortality. Grade III-IV morbidity occurred in 19% (8/42). With a median follow-up of 33.8 months after primary tumor surgery, 6/42 patients (14.3%) presented peritoneal relapse (PR). 3-year peritoneal disease free survival was 86%, with 3-year disease free survival of 78.6% and 5-year overall survival (OS) of 97.4%. Conclusion Peritoneal relapse and survival rates are remarkable in these groups of, a priori, very bad prognosis, which could suggest a beneficial effect of HIPEC. Prospective study trying to decrease peritoneal relapse in perforated or pT4 CRC. 20–30% of perforated or pT4 CRC develop peritoneal metastases in the follow-up. We find peritoneal disease at Second Look surgery before seen in the image tests. Second-Look Surgery + HIPEC decreases peritoneal relapse and gets better survival.
Collapse
Affiliation(s)
- Ángel Serrano Del Moral
- Fuenlabrada University Hospital, Camino del Molino, 2, 28942 Fuenlabrada, Madrid, Spain.,Rey Juan Carlos University, Calle Tulipán, 28933, Móstoles, Madrid, Spain
| | | | | | - Fernando Pereira Pérez
- Fuenlabrada University Hospital, Camino del Molino, 2, 28942 Fuenlabrada, Madrid, Spain.,Rey Juan Carlos University, Calle Tulipán, 28933, Móstoles, Madrid, Spain
| |
Collapse
|
47
|
Klempner SJ, Ryan DP. HIPEC for colorectal peritoneal metastases. Lancet Oncol 2021; 22:162-164. [PMID: 33476594 DOI: 10.1016/s1470-2045(20)30693-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 01/13/2023]
Affiliation(s)
- Samuel J Klempner
- Mass General Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA, USA
| | - David P Ryan
- Mass General Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
48
|
Sommariva A, Ansaloni L, Baiocchi GL, Cascinu S, Cirocchi R, Coccolini F, Deraco M, Fiorentini G, Gelmini R, Di Giorgio A, Lippolis PV, Pasqual EM, Sassaroli C, Macrì A, Sammartino P, Scaringi S, Valle M, Vaira M. Diagnostic and therapeutic algorithm for colorectal peritoneal metastases. A consensus of the peritoneal surface malignancies onco-team of the Italian society of surgical oncology. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2021; 47:164-171. [PMID: 33028502 DOI: 10.1016/j.ejso.2020.09.035] [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: 07/22/2020] [Revised: 09/04/2020] [Accepted: 09/26/2020] [Indexed: 11/30/2022]
Abstract
AIM the surgical workup for colorectal cancer peritoneal metastases (CRCPM) is complex and should be managed in specialized centers. Diagnostic and therapeutic algorithms (DTA) have been proposed to balance optimal patients management and correct use of resources. Aim of this study was to establish a consensus on DTA for CRCPM patients in Italy. METHOD a panel of 18 delegated members of centers afferent to Peritoneal Surface Malignancies Onco-team of the Italian Society of Surgical Oncology was established. A list of statements regarding the DTA of patients with CRCPM was prepared according to different activities and decision-making nodes with a defined entry and exit point. Consensus was obtained through RAND UCLA methodology. RESULTS two different DTA were defined and approved according to the modality of presentation of CRCPM (synchronous and metachronous). A consensus was also obtained on 17 of the 19 statements related to DTA. CONCLUSION a shared model of DTA is now available for healthcare providers to monitor appropriateness in diagnosis and treatment of patients with isolated peritoneal metastases from CRC.
Collapse
Affiliation(s)
- Antonio Sommariva
- Unit of Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Luca Ansaloni
- Unit of General and Emergency Surgery, Bufalini Hospital, Cesena, Italy
| | - Gian Luca Baiocchi
- Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Stefano Cascinu
- Department of Medical Oncology Vita-Salute, San Raffaele University IRCCS, Milan, Italy
| | - Roberto Cirocchi
- General Surgery and Clinical Anatomy, University of Perugia, Perugia, Italy
| | | | - Marcello Deraco
- Peritoneal Surface Malignancies Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Gianmaria Fiorentini
- Department of Onco-Hematology, Azienda Ospedaliera 'Ospedali Riuniti Marche Nord', Pesaro, Italy
| | - Roberta Gelmini
- General and Oncological Surgery Unit, AOU of Modena University of Modena and Reggio Emilia, Italy
| | - Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
| | | | - Enrico Maria Pasqual
- Advanced Surgical Oncology Unit, Department Area Medica, University of Udine, Italy
| | - Cinzia Sassaroli
- Abdominal Oncology Department, Fondazione Giovanni Pascale, IRCCS, Naples, Italy
| | - Antonio Macrì
- Department of Human Pathology, Peritoneal Surface Malignancy and Soft Tissue Sarcoma Program, University of Messina, Italy
| | - Paolo Sammartino
- Department of Surgery 'P. Valdoni', Sapienza University of Rome, Rome, Italy
| | - Stefano Scaringi
- Digestive Surgery Unit - IBD Unit, Careggi University Hospital, Florence, Italy
| | - Mario Valle
- Surgical Oncology Peritoneum and Abdomen Pathologies, National Cancer Institute "Regina Elena" Rome, Italy
| | - Marco Vaira
- Unit of Surgical Oncology, Candiolo Cancer Institute, Turin, Italy
| |
Collapse
|
49
|
Brind'Amour A, Dubé P, Tremblay JF, Soucisse ML, Mack L, Bouchard-Fortier A, McCart JA, Govindarajan A, Bischof D, Haase E, Giacomantonio C, Hebbard P, Younan R, MacNeill A, Boulanger-Gobeil C, Sidéris L. Canadian guidelines on the management of colorectal peritoneal metastases. Curr Oncol 2020; 27:e621-e631. [PMID: 33380878 PMCID: PMC7755452 DOI: 10.3747/co.27.6919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern management of colorectal cancer (crc) with peritoneal metastasis (pm) is based on a combination of cytoreductive surgery (crs), systemic chemotherapy, and hyperthermic intraperitoneal chemotherapy (hipec). Although the role of hipec has recently been questioned with respect to results from the prodige 7 trial, the role and benefit of a complete crs were confirmed, as observed with a 41-month gain in median survival in that study, and 15% of patients remaining disease-free at 5 years. Still, crc with pm is associated with a poor prognosis, and good patient selection is essential. Many questions about the optimal management approach for such patients remain, but all patients with pm from crc should be referred to, or discussed with, a pm surgical oncologist, because cure is possible. The objective of the present guideline is to offer a practical approach to the management of pm from crc and to reflect on the new practice standards set by recent publications on the topic.
Collapse
Affiliation(s)
- A Brind'Amour
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, chu de Québec-Université Laval, Quebec City
| | - P Dubé
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| | - J F Tremblay
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| | - M L Soucisse
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| | - L Mack
- Department of Surgery, University of Calgary, Calgary, AB
| | | | - J A McCart
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON
- Department of Surgery, University of Toronto, Toronto, ON
| | - A Govindarajan
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON
- Department of Surgery, University of Toronto, Toronto, ON
| | - D Bischof
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON
- Department of Surgery, University of Toronto, Toronto, ON
| | - E Haase
- Department of Surgery, University of Alberta, Edmonton, AB
| | | | - P Hebbard
- Department of Surgery, University of Manitoba, Winnipeg, MB
| | - R Younan
- Department of Surgery, University of Montreal, Montreal, QC
- Department of Surgery, Centre Hospitalier de l'Université de Montréal, Montreal, QC
| | - A MacNeill
- Department of Surgery, University of British Columbia, Vancouver, BC
| | | | - L Sidéris
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| |
Collapse
|
50
|
Gasser E, Kogler P, Lorenz A, Kafka-Ritsch R, Öfner D, Perathoner A. Do we still need CRS and HIPEC in colorectal cancer in times of modern chemotherapy and immunotherapy? MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2020; 13:430-433. [DOI: 10.1007/s12254-020-00647-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/18/2020] [Indexed: 08/30/2023]
Abstract
SummaryPeritoneal carcinomatosis from colorectal cancer is associated with a poor prognosis and is usually treated with systemic chemotherapy and immunotherapy alone. In patients with isolated peritoneal carcinomatosis (PC) without nonperitoneal metastases, however, cytoreductive surgery (CRS) has been shown to significantly improve outcome and to achieve even cure in selected patients in combination with systemic therapy. The additional use of a hyperthermic intraperitoneal chemotherapy (HIPEC) is primarily indicated to control microscopical residual tumor tissue in the peritoneal cavity after successful CRS. Another more recent option is the application of an adjuvant HIPEC to prevent peritoneal carcinomatosis in high risk patients with pT4 cancer or perforated cancer at the time of or after primary surgery. The aim of this short review is to highlight the corresponding available literature and assess the role of CRS and HIPEC in the context of modern chemotherapy and immunotherapy.
Collapse
|