1
|
Kazi M, Ajith A, Bhatt A. The Mitomycin versus Oxaliplatin debate on HIPEC in colorectal cancers - An updated systematic review and Meta-analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110080. [PMID: 40286391 DOI: 10.1016/j.ejso.2025.110080] [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: 12/25/2024] [Revised: 03/29/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Following the PRODIGE-7 trial, surgeons have shifted to the use of Mitomycin-based HIPEC from Oxaliplatin for colorectal peritoneal metastasis. While preclinical studies have demonstrated the superiority of Mitomycin over oxaliplatin, clinical studies report variable results. The objective of the meta-analysis was to determine the most efficacious drug after cytoreduction for colorectal peritoneal metastasis. METHODS he databases searched were PubMed, Cochrane Library, Scopus, CINHAL (EBSCO), and Google Scholar based on the following concepts: colorectal, peritoneal, cytoreduction, Mitomycin, and Oxaliplatin. The risk of bias was assessed using the Newcastle-Ottawa scale and certainty of the evidence was assessed using the GRADE Pro tool. The analysis was carried out using the log hazard ratio as the outcome measure for survival data. All syntheses used the Random-effects model and were reported for Oxaliplatin-based HIPEC with MMC as the reference. RESULTS Thirteen studies with 3406 patients were included in the quantitative meta-analysis. The pooled hazard ratio for overall survival was 1.03 (95 % CI: 0.786-1.349) from ten studies. Six studies reported disease-free survival and the pooled hazard ratio was 0.941 (95 % CI: 0.683-1.297). Both survival estimates had moderate statistical heterogeneity. The evidence was of very low certainty for all the outcomes due to the non-randomized nature of studies, clinical and statistical heterogeneity, serious risk of bias due to uncontrolled measured confounding, selection bias, and unequal follow-up durations. CONCLUSION Our systematic review and meta-analysis found no significant difference in survival outcomes or postoperative morbidity between MMC and Oxaliplatin-based HIPEC.
Collapse
Affiliation(s)
- Mufaddal Kazi
- Division of Gastrointestinal Surgical Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, 400012, India; Department of Surgical Oncology, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Mumbai, India.
| | - Atul Ajith
- Division of Gastrointestinal Surgical Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, 400012, India; Homi Bhabha National Institute, Mumbai, India
| | - Aditi Bhatt
- Department of Surgical Oncology, Shalby Cancer and Research Institute, Ahmedabad, 382421, India
| |
Collapse
|
2
|
Brown ZJ, Krell M, Gitlin S, Ranjbar S, Vega DA, Pawlik TM. Prospects of the surgical management of colorectal peritoneal metastasis. J Gastrointest Surg 2025; 29:101940. [PMID: 39746646 DOI: 10.1016/j.gassur.2024.101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignancy with the propensity to metastasize. Common sites of metastasis include the liver, lungs, and peritoneum, with peritoneal metastases (PMs) having the worst prognosis. Unfortunately, systemic chemotherapy is often less effective in the treatment of PMs. Therefore, removal of all visible tumor via cytoreductive surgery along with intraperitoneal (IP) therapies has been used. METHODS A comprehensive review of the literature was conducted using MEDLINE/PubMed and Web of Science with an end date of September 1, 2024, regarding cytoreductive surgery and heated IP chemotherapy for CRC PMs. RESULTS Recent studies have called into question the utility of IP chemotherapy in the treatment of CRC PMs. However, regardless of IP chemotherapy, cytoreductive surgery has demonstrated an additional survival benefit for patients with PM secondary to CRC. DISCUSSION This study reviews the pathophysiology of CRC PM, the current treatment paradigms, and a pathway for improving outcomes in patients with CRC PM.
Collapse
Affiliation(s)
- Zachary J Brown
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States.
| | - Matthew Krell
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Saige Gitlin
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Suedeh Ranjbar
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Diego Alvarez Vega
- Division of Surgical Oncology, Department of Surgery, New York University Grossman Long Island School of Medicine, New York University Langone Health, Mineola, NY, United States
| | - Timothy M Pawlik
- Division of Surgical Oncology, Department of Surgery, Wexner Medical Center and James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
3
|
Alaswad MM, Arabi TZ, Alshahrani AS, Sabbah BN, Jaamour HW, AlOtry AA, Saleh TW, Alhosainy ZH, Elkordy FH, Elnegali AI, Tlayjeh MH, Kseibi FE, Azzam AZ, Amin TM. Cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy and intraoperative radiation therapy in the management of gastric cancer: a 10-year single center experience. World J Surg Oncol 2025; 23:62. [PMID: 39987135 PMCID: PMC11846258 DOI: 10.1186/s12957-025-03678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/19/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The rationale behind the use of HIPEC involves targeted elimination of microscopic peritoneal metastasis, a common route for GCa dissemination, thereby improving the overall survival and reducing recurrences. Moreover, the reasoning behind the use of IORT is enhanced loco-regional control and, therefore, reducing recurrence rates. METHODS From February 2013 to June 2023, all GCa patients who underwent HIPEC plus IORT during surgery were included in this study. Median overall survival (OS) and disease-free (DFS) survival were used to evaluate the efficacy of this treatment strategy amongst GCa patients, along with the rate of occurrence and severity of post-operative complications associated with this treatment strategy. RESULTS The median OS and DFS were 63 and 87 months, respectively. More than one-third of the patients in our cohort did not develop any post-operative complications. In patients who developed post-operative complications, the median number of post-operative complications was 1 (IQR 1-2). Most encountered complications were Clavien-Dindo (CD) grade II complications (33.33%) and no in-hospital mortality was observed. CONCLUSIONS This complex, multimodal treatment strategy results in a significantly prolonged OS and DFS when compared to other treatment strategies for gastric cancer patients, with no added morbidity or mortality.
Collapse
Affiliation(s)
- Marwan M Alaswad
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia.
| | - Tarek Z Arabi
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Asma S Alshahrani
- Department of Population Health, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Belal N Sabbah
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Heba W Jaamour
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Abdullah A AlOtry
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Tariq W Saleh
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Ziad H Alhosainy
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Fadwa H Elkordy
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Aya I Elnegali
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Mohamed H Tlayjeh
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Firas E Kseibi
- College of Medicine, Alfaisal University, Al Zahrawai Street, Riyadh, Saudi Arabia
| | - Ayman Z Azzam
- Department of Surgical Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Department of General Surgery, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Tarek M Amin
- Department of Surgical Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Lai C, Liu J, Zhou J, Zhou H. The first-line antihypertensive nitrendipine potentiated the therapeutic effect of oxaliplatin by downregulating CACNA1D in colorectal cancer. Open Med (Wars) 2025; 20:20241138. [PMID: 39958978 PMCID: PMC11826243 DOI: 10.1515/med-2024-1138] [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: 05/20/2024] [Revised: 10/10/2024] [Accepted: 12/24/2024] [Indexed: 02/18/2025] Open
Abstract
Background Oxaliplatin (OXA) is among the most common chemotherapy drugs and is the base component of the FOLFOX regimen (OXA + leucovorin + 5-fluorouracil) and CapeOX regimen (OXA + capecitabine). Resistance to and failure of these two OXA-based regimens often results in poor outcomes in patients with colorectal cancer (CRC). Nitrendipine (NTD) is a first-line antihypertensive drug commonly used in hypertension and coronary heart disease with confirmed low toxicity and side effects. However, the potential benefits of NTD for CRC progression and therapy remain unclear. Methods Cell counting kit-8 (CCK-8) detection, colony formation assay, wound-healing assay, Transwell assay, SynergyFinder webtool, and subcutaneous tumor models were used to assess the effect of NTD with OXA on CRC inhibition in vitro and in vivo. Bioinformatics tools including Human Protein Atlas (HPA), quantitative real-time polymerase chain reaction, western blotting analyses, lentivirus transfection, and rescue experiment were used to investigate the mechanism(s) of the related action. Results Utilizing murine and human CRC cell lines, the in vitro and in vivo experiment demonstrated that NTD inhibited cell proliferation, migration, and invasion, and the synergy scores calculated by SynergyFinder indicated that NTD exhibited synergistic activity with the chemotherapeutic drug OXA. The CCK-8 detection, animal model, and rescue experiment results demonstrated that NTD suppressed CRC progression and potentiated OXA therapeutic effect by downregulating calcium voltage-gated channel subunit alpha1 D (CACNA1D). Conclusions This study presents novel data on first-line antihypertensive NTD, exerting inhibitory effects on cell proliferation and migration in CRC and revealing synergistic activity with OXA by downregulating CACNA1D. NTD may be a candidate as a promising chemosensitizer as an OXA new combination to improve the efficacy and safety of CRC therapy.
Collapse
Affiliation(s)
- Chengzhe Lai
- Department of Cardiology, The Fourth Affiliated Hospital of Guangzhou Medical University, Zengcheng, Guangzhou, China
| | - Jinghu Liu
- Department of Cardiology, The Fourth Affiliated Hospital of Guangzhou Medical University, Zengcheng, Guangzhou, China
| | - Jingna Zhou
- Department of Medicine, Taizhou University, Zhejiang, China
| | - Haokun Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Guangzhou Medical University, Zengcheng, Guangzhou, China
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Moon KS, Song JM, Yi J, Pham QK, Ahn SH. Determination of mitomycin C in rat plasma by liquid chromatography-tandem mass spectrometry and its application for determining pharmacokinetics in rat. Heliyon 2024; 10:e32927. [PMID: 38988565 PMCID: PMC11233998 DOI: 10.1016/j.heliyon.2024.e32927] [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: 03/12/2024] [Revised: 05/10/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
To develop the liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for measuring mitomycin C in rat plasma, samples were processed using solid-phase extraction, with the internal standard being carbamazepine. A reversed phased C18 column was utilized for the LC-MS/MS study, and mobile phases consisting of 0.1 % formic acid in acetonitrile and water were injected into it at a rate of 0.3 mL/min. Multiple reaction monitoring in positive-ion mode with precursor-product ion pairs 335.3 → 242.3 (mitomycin C) and 237.1 → 194.1 (carbamazepine) was employed to quantify the compounds. The linear range in plasma was found to be 10-4000 ng/mL (r2 = 0.992). The inter-batch and intra-batch precision were <14.3 % (LLOQ: 14.7 %) and 13.4 % (LLOQ: 16.1 %), respectively. The recovery and the matrix effect of mitomycin C in plasma were 113 % and 111 %, respectively. Mitomycin C was stable under the conditions of this assay method. In the end, this approach proved effective in a pharmacokinetic investigation with the intravenous and oral administration of mitomycin C to rats.
Collapse
Affiliation(s)
- Kyung-Sun Moon
- Laboratory of Pharmaceutical Sciences, College of Pharmacy, Kangwon National University, Chuncheon, Gangwondo, 24341, Republic of Korea
| | - Jong-Min Song
- Laboratory of Pharmaceutical Sciences, College of Pharmacy, Kangwon National University, Chuncheon, Gangwondo, 24341, Republic of Korea
| | - JiMin Yi
- Laboratory of Pharmaceutical Sciences, College of Pharmacy, Kangwon National University, Chuncheon, Gangwondo, 24341, Republic of Korea
| | - Quynh Khoa Pham
- Laboratory of Pharmaceutical Sciences, College of Pharmacy, Kangwon National University, Chuncheon, Gangwondo, 24341, Republic of Korea
| | - Sung-Hoon Ahn
- Laboratory of Pharmaceutical Sciences, College of Pharmacy, Kangwon National University, Chuncheon, Gangwondo, 24341, Republic of Korea
| |
Collapse
|
8
|
Ballal DS, Saklani AP. HIPECT4 Trial-the Devil Is in the Details! Is Prophylactic HIPEC and Cytoreduction the New Standard of Care for T4 Colorectal Cancer? Indian J Surg Oncol 2024; 15:302-303. [PMID: 38741653 PMCID: PMC11088578 DOI: 10.1007/s13193-024-01899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/06/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Devesh S. Ballal
- Division of Colo-Rectal and Peritoneal Surface Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Room 1212, 12 Floor Homi Babha Block, Dr E. Borges Marg, Parel, Mumbai, 400012 India
| | - Avanish P. Saklani
- Division of Colo-Rectal and Peritoneal Surface Oncology, Department of Surgical Oncology, Tata Memorial Hospital, Room 1212, 12 Floor Homi Babha Block, Dr E. Borges Marg, Parel, Mumbai, 400012 India
| |
Collapse
|
9
|
Gladieff L. [HIPEC morbidity and implications for post-surgical treatment. A medical oncologist advice]. Bull Cancer 2024; 111:248-253. [PMID: 36822957 DOI: 10.1016/j.bulcan.2023.01.011] [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: 10/25/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/23/2023]
Abstract
The fear that the medical oncologist may have is that HIPEC integrated into a multidisciplinary care pathway will negatively impact the treatments that will follow. This fear is largely related to the side effects, which are themselves dependent on the medication used. Cisplatin, most frequently used for epithelial ovarian cancers, has essentially renal toxicity, which can be avoided by the use of sodium thiosulfate. Oxaliplatin induces more severe toxicities post surgery than mitomycin C in colorectal cancers. However, the data from randomized trials are reassuring for the medical oncologist concerning the course of postoperative treatment, as long as HIPEC is performed according to a standardized protocol, within trained teams, and after multidisciplinary discussion concerning its modalities.
Collapse
Affiliation(s)
- Laurence Gladieff
- Institut Claudius Regaud IUCT-Oncopole, département d'oncologie médicale, 1, avenue Irène Joliot-Curie, 31059 Toulouse cedex, France.
| |
Collapse
|
10
|
Lukácsi S, Munkácsy G, Győrffy B. Harnessing Hyperthermia: Molecular, Cellular, and Immunological Insights for Enhanced Anticancer Therapies. Integr Cancer Ther 2024; 23:15347354241242094. [PMID: 38818970 PMCID: PMC11143831 DOI: 10.1177/15347354241242094] [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: 12/28/2023] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 06/01/2024] Open
Abstract
Hyperthermia, the raising of tumor temperature (≥39°C), holds great promise as an adjuvant treatment for cancer therapy. This review focuses on 2 key aspects of hyperthermia: its molecular and cellular effects and its impact on the immune system. Hyperthermia has profound effects on critical biological processes. Increased temperatures inhibit DNA repair enzymes, making cancer cells more sensitive to chemotherapy and radiation. Elevated temperatures also induce cell cycle arrest and trigger apoptotic pathways. Furthermore, hyperthermia modifies the expression of heat shock proteins, which play vital roles in cancer therapy, including enhancing immune responses. Hyperthermic treatments also have a significant impact on the body's immune response against tumors, potentially improving the efficacy of immune checkpoint inhibitors. Mild systemic hyperthermia (39°C-41°C) mimics fever, activating immune cells and raising metabolic rates. Intense heat above 50°C can release tumor antigens, enhancing immune reactions. Using photothermal nanoparticles for targeted heating and drug delivery can also modulate the immune response. Hyperthermia emerges as a cost-effective and well-tolerated adjuvant therapy when integrated with immunotherapy. This comprehensive review serves as a valuable resource for the selection of patient-specific treatments and the guidance of future experimental studies.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Gyöngyi Munkácsy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
- University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| |
Collapse
|
11
|
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
|
12
|
Saklani A, Kazi M, Desouza A, Sharma A, Engineer R, Krishnatry R, Gudi S, Ostwal V, Ramaswamy A, Dhanwat A, Bhargava P, Mehta S, Sundaram S, Kale A, Goel M, Patkar S, Vartey G, Kulkarni S, Baheti A, Ankathi S, Haria P, Katdare A, Choudhari A, Ramadwar M, Menon M, Patil P. Tata Memorial Centre Evidence Based Management of Colorectal cancer. Indian J Cancer 2024; 61:S29-S51. [PMID: 38424681 DOI: 10.4103/ijc.ijc_66_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
This review article examines the evidence-based management of colorectal cancers, focusing on topics characterized by ongoing debates and evolving evidence. To contribute to the scientific discourse, we intentionally exclude subjects with established guidelines, concentrating instead on areas where the current understanding is dynamic. Our analysis encompasses a thorough exploration of critical themes, including the evidence surrounding complete mesocolic excision and D3 lymphadenectomy in colon cancers. Additionally, we delve into the evolving landscape of perioperative chemotherapy in both colon and rectal cancers, considering its nuanced role in the context of contemporary treatment strategies. Advancements in surgical techniques are a pivotal aspect of our discussion, with an emphasis on the utilization of minimally invasive approaches such as laparoscopy and robotic surgery in both colon and rectal cancers, including advanced rectal cases. Moving beyond conventional radical procedures, we scrutinize the feasibility and implications of endoscopic resections for small tumors, explore the paradigm of organ preservation in locally advanced rectal cancers, and assess the utility of total neoadjuvant therapy in the current treatment landscape. Our final segment reviews pivotal trials that have significantly influenced the management of colorectal liver and peritoneal metastasis.
Collapse
Affiliation(s)
- Avanish Saklani
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Mufaddal Kazi
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
- Department of Surgical Oncology, Advanced Centre of the Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, India
| | - Ashwin Desouza
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Ankit Sharma
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
- Department of Surgical Oncology, Advanced Centre of the Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, India
| | - Reena Engineer
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, India
| | - Rahul Krishnatry
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, India
| | - Shivkumar Gudi
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, India
| | - Vikas Ostwal
- Homi Bhabha National Institute, Mumbai, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Anant Ramaswamy
- Homi Bhabha National Institute, Mumbai, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Aditya Dhanwat
- Homi Bhabha National Institute, Mumbai, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Prabhat Bhargava
- Homi Bhabha National Institute, Mumbai, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Shaesta Mehta
- Homi Bhabha National Institute, Mumbai, India
- Department of Digestive Diseases and Clinical Nutrition, Tata Memorial Hospital, Mumbai, India
| | - Sridhar Sundaram
- Homi Bhabha National Institute, Mumbai, India
- Department of Digestive Diseases and Clinical Nutrition, Tata Memorial Hospital, Mumbai, India
| | - Aditya Kale
- Homi Bhabha National Institute, Mumbai, India
- Department of Digestive Diseases and Clinical Nutrition, Tata Memorial Hospital, Mumbai, India
| | - Mahesh Goel
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Shraddha Patkar
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Gurudutt Vartey
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Suyash Kulkarni
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, India
| | - Akshay Baheti
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, India
| | - Suman Ankathi
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, India
| | - Purvi Haria
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, India
| | - Aparna Katdare
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, India
| | - Amit Choudhari
- Homi Bhabha National Institute, Mumbai, India
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, India
| | - Mukta Ramadwar
- Homi Bhabha National Institute, Mumbai, India
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Munita Menon
- Homi Bhabha National Institute, Mumbai, India
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Prachi Patil
- Homi Bhabha National Institute, Mumbai, India
- Department of Digestive Diseases and Clinical Nutrition, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
13
|
Hou Z, Qiu G, Xie Q, Jin Z, Mi S, Huang J. The prophylactic role of mitomycin C-based hyperthermic intraperitoneal chemotherapy (MMC-based HIPEC) on peritoneal metastasis of spontaneously ruptured hepatocellular carcinoma (srHCC): A pilot study. Glob Health Med 2023; 5:336-344. [PMID: 38162434 PMCID: PMC10730926 DOI: 10.35772/ghm.2023.01081] [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: 07/20/2023] [Revised: 09/29/2023] [Accepted: 10/07/2023] [Indexed: 01/03/2024]
Abstract
Hepatocellular carcinoma (HCC) was featured as spontaneous rupture hemorrhage under intratumoral overpressure. Spontaneous rupture hepatocellular carcinoma (srHCC) has a high propensity for peritoneal metastasis (PM). Although HIPEC has become standard treatment for malignancies with PM, it has been poorly described in srHCC. We conducted a single-arm, open-label, single-center, prospective study to explore the prophylactic role of MMC-based HIPEC on PM of srHCC. A total of 7 patients were collected from April 1, 2021 to April 30, 2022. HIPEC was conducted 3 times on the first, third and fifth postoperative days. 15 mg/m2 of MMC was used with 60 minutes perfusion at 43°C. The primary end-point was local peritoneum recurrence free survival (RFS), whereas the secondary end-point was systemic RFS and overall survival (OS). The mean hepatectomy operation time was 232 minutes (SD: 124.08 minutes). The median bleeding loss was 200 mL (range 50-400 mL). The mean hospital stay was 13 days (SD: 3.42 days). Only mild abdominal distension was reported in 4 patients (57%). There were no patients who suffered from life-threatening intra-abdominal and extra-abdominal complications (EAC). At the data cut-off (April 30, 2023), one patient (14%) had died due to cachexia. Local peritoneal recurrence occurred in three patients (43%). Median follow-up was 16.1 months (IQR: 12.8-16.6 months). Median local peritoneum RFS was 12.3 months (95% CI: 7.0- 17.5; 4 events) and median overall RFS was 7.5 months (95% CI: 4.2-10.8; 6 events). MMC-based HIPEC was safe and feasible in selected patients of srHCC. It showed a positive tendency in preventing PM, but large-scale research should be continued.
Collapse
Affiliation(s)
- Ziqi Hou
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Guoteng Qiu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qingyun Xie
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhaoxing Jin
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shizheng Mi
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Vallejo JSA, Queiroz FLDE, Lacerda Filho A, França Neto PR, Costa BXMDA, Paiva RA, Garcia SLM, Silva SB. Assessing morbidity, mortality, and survival in patients with peritoneal carcinomatosis undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Rev Col Bras Cir 2023; 50:e20233421. [PMID: 37075463 PMCID: PMC10508679 DOI: 10.1590/0100-6991e-20233421-en] [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: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 04/21/2023] Open
Abstract
OBJECTIVE Peritoneal carcinomatosis (PC) indicates advanced stage cancer, which is generally associated with a poor outcome and a 6 to 12 months. Cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) is an option for treating patients with primary PC, such as mesothelioma, or secondary PC, such as colorectal cancer (CRC) or pseudomixoma. Until recently, such patients were deemed untreatable. The purpose of this study was to assess the results of CRS + HIPEC in patients with PC. Postoperative complications, mortality and survival rates were evaluated according to the diagnosis. RESULTS Fifty-six patients with PC, undergoing full CRS + HIPEC between October 2004 and January 2020, were enrolled. The mortality rate was 3.8% and the morbidity rate was 61.5%. Complications were significantly higher in proportion to the duration of surgery (p<0.001). The overall survival rates, as shown in the Kaplan-Meyer curve, were respectively 81%, 74% and 53% at 12, 24 and 60 months. Survival rates according to each diagnosis for the same periods were 87%, 82% and 47% in patients with pseudomixoma, and 77%, 72% and 57% in patients with CRC (log-rank 0.371, p=0.543). CONCLUSION CRS with HIPEC is an option for pacients with primary or secondary PC. Although complication rates are high, a longer survival rate may be attained compared to those seen in previously published results; in some cases, patients may even be cured.
Collapse
|
15
|
Mangieri CW, Valenzuela CD, Solsky IB, Erali RA, Votanopoulos KI, Shen P, Levine EA. Exposure to Neoadjuvant Oxaliplatin-Containing Chemotherapy, Does it Effect Intraperitoneal Hyperthermic Chemotherapy Perfusion? Ann Surg Oncol 2023; 30:2486-2493. [PMID: 36484904 DOI: 10.1245/s10434-022-12933-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Patients undergoing cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS/HIPEC) are commonly exposed to oxaliplatin neoadjuvant chemotherapy (NAT) regimens. The impact of systemic exposure to oxaliplatin prior to HIPEC with oxaliplatin is unknown. METHODS We conducted a retrospective review of our institutional registry of CRS/HIPEC cases who received oxaliplatin-containing NAT, and compared patients who underwent HIPEC with oxaliplatin versus cases perfused with mitomycin C. The primary outcome was survival, defined by overall survival (OS) and disease-free survival (DFS). Subgroup analysis was performed based on primary tumor etiology and completeness of cytoreduction. RESULTS A total of 333 cases satisfied the selection criteria-159 appendiceal primaries (all high-grade disease) and 174 colorectal cases. Thirty-one cases (9.3%) underwent HIPEC with oxaliplatin, with the remaining 302 cases (90.7%) receiving mitomycin C. Both cohorts were identical in regard to baseline characteristics, and both groups were alike in regard to NAT regimens and oxaliplatin exposure. There was no difference in survival outcomes. OS times were 2.9 (± 2.8) and 2.8 ( ± 3.6) years for oxaliplatin and mitomycin C perfusions, respectively (p = 0.94), and the 5-year OS rates were also similar at 9.7 and 18.5% (odds ratio [OR] 0.49, 95% confidence interval [CI] 0.14-1.67, p = 0.24) for oxaliplatin and mitomycin cases, respectively. Likewise, DFS findings were similar, with survival of 2.5 (± 4.5) and 1.8 (± 2.4) years for oxaliplatin and mitomycin perfusions, respectively (p = 0.21). There was no difference in 5-year DFS rates, at 10.5 and 7.8% (OR 1.39, 95% CI 0.30-6.56, p = 0.68) for oxaliplatin and mitomycin C, respectively. Subgroup analysis found minimal discordant findings from the main results. CONCLUSION This analysis found no discernable association with NAT oxaliplatin exposure in regard to survival outcomes following CRS/HIPEC stratified out by perfusion agent.
Collapse
Affiliation(s)
- Christopher W Mangieri
- Division of Surgical Oncology, Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | - Cristian D Valenzuela
- Division of Surgical Oncology, Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | - Ian B Solsky
- Division of Surgical Oncology, Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | - Richard A Erali
- Division of Surgical Oncology, Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | | | - Perry Shen
- Division of Surgical Oncology, Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | - Edward A Levine
- Division of Surgical Oncology, Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA.
| |
Collapse
|
16
|
Frech FS, Hernandez L, Urbonas R, Zaken GA, Dreyfuss I, Nouri K. Hypertrophic Scars and Keloids: Advances in Treatment and Review of Established Therapies. Am J Clin Dermatol 2023; 24:225-245. [PMID: 36662366 DOI: 10.1007/s40257-022-00744-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 01/21/2023]
Abstract
Hypertrophic scars and keloids can have significant detrimental effects on patients both psychosocially and functionally. A careful identification of patient risk factors and a comprehensive management plan are necessary to optimize outcomes. Patients with a history of dystrophic scarring should avoid unnecessary procedures and enhance the wound-healing process using various preventive strategies. As there is no single, fully efficacious treatment modality, prevention remains the best approach in reducing aberrant scar formation. When prevention therapies fail, keloids have been shown to be respond to a variety of therapies including topical and injectable corticosteroids, 5-fluorouracil, radiotherapy, lasers, and surgical excision, all with varying efficacies. As such, management should be tailored to the individual patient's risk factors with the use of combination therapies to reduce recurrence rates. Still, keloid and hypertrophic scar therapies are widely diverse with novel treatment modalities providing alternatives for recurring lesions. Laser-assisted drug delivery, skin priming, and novel topical therapies may provide alternative options for the management of hypertrophic scars and keloids.
Collapse
Affiliation(s)
- Fabio Stefano Frech
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Miami, FL, 33136, USA.
| | - Loren Hernandez
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Miami, FL, 33136, USA
| | - Rebecca Urbonas
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Ghufran Abo Zaken
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Miami, FL, 33136, USA
| | - Isabella Dreyfuss
- Nova Southeastern University, Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, FL, USA
| | - Keyvan Nouri
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Miami, FL, 33136, USA
| |
Collapse
|
17
|
Choi M, Harper MM, Pandalai PK, Abdel-Misih SRZ, Patel RA, Ellis CS, Reusch E, Reynolds J, Vacchi-Suzzi C, Park JM, Georgakis GV, Kim J. A Multicenter Phase 1 Trial Evaluating Nanoliposomal Irinotecan for Heated Intraperitoneal Chemotherapy Combined with Cytoreductive Surgery for Patients with Peritoneal Surface Disease. Ann Surg Oncol 2023; 30:804-813. [PMID: 36344711 DOI: 10.1245/s10434-022-12723-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Nanoliposomal irinotecan (nal-IRI) is a promising novel hyperthermic intraperitoneal chemotherapy (HIPEC) agent given its enhanced efficacy against gastrointestinal tumors, safety profile, thermo-synergy, and heat stability. This report describes the first in-human phase 1 clinical trial of nal-IRI during cytoreductive surgery (CRS) and HIPEC. METHODS Patients with peritoneal surface disease (PSD) from appendiceal and colorectal neoplasms were enrolled in a 3 + 3 dose-escalation trial using nal-IRI (70-280 mg/m2) during HIPEC for 30 min at 41 ± 1 °C. The primary outcome was safety. The secondary outcomes were pharmacokinetics (PK) and disease-free survival. Adverse events (AEs) categorized as grade 2 or higher were recorded. The serious AEs (SAEs) were mortality, grade ≥ 3 AEs, and dose-limiting toxicity (DLT). Irinotecan and active metabolite SN38 were measured in plasma and peritoneal washings. RESULTS The study enrolled 18 patients, who received nal-IRI during HIPEC at 70 mg/m2 (n = 3), 140 mg/m2 (n = 6), 210 mg/m2 (n = 3), and 280 mg/m2 (n = 6). No DLT or mortality occurred. The overall morbidity for CRS/HIPEC was 39% (n = 7). Although one patient experienced neutropenia, no AE (n = 131) or SAE (n = 3) was definitively attributable to nal-IRI. At 280 mg/m2, plasma irinotecan and SN38 measurements showed maximum concentrations of 0.4 ± 0.6 µg/mL and 3.0 ± 2.4 ng/mL, a median time to maximum concentration of 24.5 and 26 h, and areas under the curve of 22.6 h*µg/mL and 168 h*ng/mL, respectively. At the 6-month follow-up visit, 83% (n = 15) of the patients remained disease-free. CONCLUSIONS In this phase 1 HIPEC trial (NCT04088786), nal-IRI was observed to be safe, and PK profiling showed low systemic absorption overall. These data support future studies testing the efficacy of nal-IRI in CRS/HIPEC.
Collapse
Affiliation(s)
- Minsig Choi
- Department of Hematology and Oncology, Stony Brook University, Stony Brook, NY, USA
| | - Megan M Harper
- Division of Surgical Oncology, University of Kentucky, Lexington, KY, USA
| | - Prakash K Pandalai
- Division of Surgical Oncology, University of Kentucky, Lexington, KY, USA
| | | | - Reema A Patel
- Division of Hematology and Oncology, University of Kentucky, Lexington, KY, USA
| | | | - Ellen Reusch
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jeri Reynolds
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | | | - Jinha M Park
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | | | - Joseph Kim
- Division of Surgical Oncology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
18
|
Sommariva A, Tonello M, Coccolini F, De Manzoni G, Delrio P, Pizzolato E, Gelmini R, Serra F, Rreka E, Pasqual EM, Marano L, Biacchi D, Carboni F, Kusamura S, Sammartino P. Colorectal Cancer with Peritoneal Metastases: The Impact of the Results of PROPHYLOCHIP, COLOPEC, and PRODIGE 7 Trials on Peritoneal Disease Management. Cancers (Basel) 2022; 15:165. [PMID: 36612161 PMCID: PMC9818482 DOI: 10.3390/cancers15010165] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
HIPEC is a potentially useful locoregional treatment combined with cytoreduction in patients with peritoneal colorectal metastases. Despite being widely used in several cancer centers around the world, its role had never been investigated before the results of three important RCTs appeared on this topic. The PRODIGE 7 trial clarified the role of oxaliplatin-based HIPEC in patients treated with radical surgery. Conversely, the PROPHYLOCHIP and the COLOPEC were designed to chair the role of HIPEC in patients at high risk of developing peritoneal metastases. Although all three trials demonstrated the relative ineffectiveness of HIPEC for treating or preventing peritoneal metastases, these results are not sufficient to abandon this technique. In addition to some criticisms relating to the design of the trials and their statistical value, the oxaliplatin-based HIPEC was found to be ineffective in preventing or treating peritoneal colorectal metastases, especially in patients already treated with systemic platinum-based chemotherapy. Several studies are ongoing investigating further HIPEC drugs and regimens. The review deeply discussed all the aspects and relapses of this new evidence.
Collapse
Affiliation(s)
- Antonio Sommariva
- Advanced Surgical Oncology Unit, Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Marco Tonello
- Advanced Surgical Oncology Unit, Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Federico Coccolini
- General, Emergency and Trauma Surgery, Pisa University Hospital, 56124 Pisa, Italy
| | | | - Paolo Delrio
- Colorectal Surgical Oncology, Abdominal Oncology Department, “Fondazione Giovanni Pascale” IRCCS, 80131 Naples, Italy
| | - Elisa Pizzolato
- Advanced Surgical Oncology Unit, Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Roberta Gelmini
- SC Chirurgia Generale d’Urgenza ed Oncologica, AOU Policlinico di Modena, 41124 Modena, Italy
| | - Francesco Serra
- SC Chirurgia Generale d’Urgenza ed Oncologica, AOU Policlinico di Modena, 41124 Modena, Italy
| | - Erion Rreka
- General and Peritoneal Surgery, Department of Surgery, Pisa University Hospital, 56124 Pisa, Italy
| | - Enrico Maria Pasqual
- DAME University of Udine-AOUD Center Advanced Surgical Oncology, 33100 Udine, Italy
| | - Luigi Marano
- Department of Medicine, Surgery, and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, 53100 Siena, Italy
| | - Daniele Biacchi
- CRS and HIPEC Unit, Pietro Valdoni, Umberto I Policlinico di Roma, 00161 Roma, Italy
| | - Fabio Carboni
- Peritoneal Tumours Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Shigeki Kusamura
- Peritoneal Surface Malignancies Unit, Fondazione Istituto Nazionale Tumori IRCCS, 20133 Milano, Italy
| | - Paolo Sammartino
- CRS and HIPEC Unit, Pietro Valdoni, Umberto I Policlinico di Roma, 00161 Roma, Italy
| |
Collapse
|
19
|
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
|
20
|
Peeters H, van Zwol EM, Brancato L, M C da Cunha MG, Bogers J. Systematic review of the registered clinical trials for oncological hyperthermia treatment. Int J Hyperthermia 2022; 39:806-812. [PMID: 35710344 DOI: 10.1080/02656736.2022.2076292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND The use of heat to treat various diseases is called hyperthermia treatment (HT). Since the 1970s, the anti-cancer effects of HT have been investigated. Different HT techniques can be categorized as local, regional and whole-body hyperthermia treatment (WBHT). We aim to provide a summary of recent research done on HT to treat cancer. METHODS In July 2020 ClinicalTrials.gov were systematically searched for all trials including hyperthermia and cancer registered between 2000 and 2020. Studies were excluded when they did not concern hyperthermal treatment, when they were not oncological studies, when they were observational or other non-interventional studies. RESULTS Of 1654 identified trials, 235 were included. Of these 235 studies, 123 described the use of HIPEC (52.3%), 44 other types of regional HT (18.7%), 45 local HT (19.1%) and 15 WBHT (6.4%). A steady increase (720%) in research to hyperthermic intraperitoneal chemotherapy (HIPEC) can be observed in the last decade. Although HIPEC is the most researched HT modality, an evolution in other HT technologies could be observed during the past decade. CONCLUSIONS Research to HT to treat cancer has expanded fast. Some techniques, for example HIPEC start to be used outside of research context, but overall, more research is needed to establish a clear effect of these HT techniques.
Collapse
Affiliation(s)
| | | | | | | | - J Bogers
- ElmediX NV, Mechelen, Belgium.,Laboratory for Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
21
|
Current Trends in Cytoreductive Surgery (CRS) and Hyperthermic Intraperitoneal Chemotherapy (HIPEC) for Peritoneal Disease from Appendiceal and Colorectal Malignancies. J Clin Med 2022; 11:jcm11102840. [PMID: 35628966 PMCID: PMC9143396 DOI: 10.3390/jcm11102840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/10/2022] Open
Abstract
Peritoneal carcinomatosis (PC) is a poor prognostic factor for all malignancies. This extent of metastatic disease progression remains difficult to treat with systemic therapies due to poor peritoneal vascularization resulting in limited drug delivery and penetration into tissues. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are surgical interventions that directly target peritoneal tumors and have improved outcomes for PC resulting from appendiceal and colorectal cancer (CRC). Despite these radical therapies, long-term survival remains infrequent, and recurrence is common. The reasons for these outcomes are multifactorial and signal the need for the continued development of novel therapeutics, techniques, and approaches to improve outcomes for these patients. Here, we review landmark historical studies that serve as the foundation for current recommendations, recent discoveries, clinical trials, active research, and areas of future interest in CRS/HIPEC to treat PC originating from appendiceal and colorectal malignancies.
Collapse
|
22
|
Management of Peritoneal Disease in Colorectal Cancer. Hematol Oncol Clin North Am 2022; 36:569-582. [DOI: 10.1016/j.hoc.2022.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
23
|
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
|
24
|
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
|