1
|
Li X, Liu G, Wu W. Progress in Biological Research and Treatment of Pseudomyxoma Peritonei. Cancers (Basel) 2024; 16:1406. [PMID: 38611084 PMCID: PMC11010892 DOI: 10.3390/cancers16071406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Pseudomyxoma peritonei (PMP) is a rare disease characterized by extensive peritoneal implantation and mass secretion of mucus after primary mucinous tumors of the appendix or other organ ruptures. Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is currently the preferred treatment, with excellent efficacy and safety, and is associated with breakthrough progress in long-term disease control and prolonged survival. However, the high recurrence rate of PMP is the key challenge in its treatment, which limits the clinical application of multiple rounds of CRS-HIPEC and does not benefit from conventional systemic chemotherapy. Therefore, the development of alternative therapies for patients with refractory or relapsing PMP is critical. The literature related to PMP research progress and treatment was searched in the Web of Science, PubMed, and Google Scholar databases, and a literature review was conducted. The overview of the biological research, treatment status, potential therapeutic strategies, current research limitations, and future directions associated with PMP are presented, focuses on CRS-HIPEC therapy and alternative or combination therapy strategies, and emphasizes the clinical transformation prospects of potential therapeutic strategies such as mucolytic agents and targeted therapy. It provides a theoretical reference for the treatment of PMP and the main directions for future research.
Collapse
Affiliation(s)
- Xi Li
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guodong Liu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
2
|
Chia DKA, Demuytere J, Ernst S, Salavati H, Ceelen W. Effects of Hyperthermia and Hyperthermic Intraperitoneal Chemoperfusion on the Peritoneal and Tumor Immune Contexture. Cancers (Basel) 2023; 15:4314. [PMID: 37686590 PMCID: PMC10486595 DOI: 10.3390/cancers15174314] [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: 07/22/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Hyperthermia combined with intraperitoneal (IP) drug delivery is increasingly used in the treatment of peritoneal metastases (PM). Hyperthermia enhances tumor perfusion and increases drug penetration after IP delivery. The peritoneum is increasingly recognized as an immune-privileged organ with its own distinct immune microenvironment. Here, we review the immune landscape of the healthy peritoneal cavity and immune contexture of peritoneal metastases. Next, we review the potential benefits and unwanted tumor-promoting effects of hyperthermia and the associated heat shock response on the tumor immune microenvironment. We highlight the potential modulating effect of hyperthermia on the biomechanical properties of tumor tissue and the consequences for immune cell infiltration. Data from translational and clinical studies are reviewed. We conclude that (mild) hyperthermia and HIPEC have the potential to enhance antitumor immunity, but detailed further studies are required to distinguish beneficial from tumor-promoting effects.
Collapse
Affiliation(s)
- Daryl K. A. Chia
- Department of Surgery, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Jesse Demuytere
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Sam Ernst
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Hooman Salavati
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
3
|
Ramirez MF, Guerra-Londono JJ, Owusu-Agyemang P, Fournier K, Guerra-Londono CE. Temperature management during cytoreductive surgery with hyperthermic intraperitoneal chemotherapy. Front Oncol 2023; 12:1062158. [PMID: 36741691 PMCID: PMC9894316 DOI: 10.3389/fonc.2022.1062158] [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: 10/10/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
In addition to attaining complete or near complete cytoreduction, the instillation of select heated chemotherapeutic agents into the abdominal cavity has offered a chance for cure or longer survival inpatients with peritoneal surface malignancies. While the heating of chemotherapeutic agents enhances cytotoxicity, the resulting systemic hyperthermia has been associated with an increased risk of severe hyperthermia and its associated complications. Factors that have been associated with an increased risk of severe hyperthermia include intraoperative blood transfusions and longer perfusion duration. However, the development of severe hyperthermia still remains largely unpredictable. Thus, at several institutions, cooling protocols are employed during cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS-HIPEC). Cooling protocols for CRS-HIPEC are not standardized and may be associated with episodes of severe hyperthermia or alternatively hypothermia. In theory, excessive cooling could result in a decreased effectiveness of the intraperitoneal chemotherapeutic agents. This presumption has been supported by a recent study of 214 adults undergoing CRS-HIPEC, where failure to attain a temperature of 38° C at the end of chemo-perfusion was associated with worse survival. Although not statistically significant, failure to maintain a temperature of 38° C for at least 30 minutes was associated with worse survival. Although studies are limited in this regard, the importance of maintaining a steady state of temperature during the hyperthermic phase of intraperitoneal chemotherapy administration cannot be disregarded. The following article describes the processes and physiological mechanisms responsible for hyperthermia during CRS-HIPEC. The challenges associated with temperature management during CRS-HIPEC and methods to avoid severe hypothermia and hyperthermia are also described.
Collapse
Affiliation(s)
- Maria F. Ramirez
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States,*Correspondence: Maria F. Ramirez,
| | - Juan Jose Guerra-Londono
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pascal Owusu-Agyemang
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Keith Fournier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Carlos E. Guerra-Londono
- Department of Anesthesiology, Pain Management, & Perioperative Medicine, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
4
|
Hu J, Wang Z, Wang X, Xie S. Side-effects of hyperthermic intraperitoneal chemotherapy in patients with gastrointestinal cancers. PeerJ 2023; 11:e15277. [PMID: 37138820 PMCID: PMC10150720 DOI: 10.7717/peerj.15277] [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: 11/09/2022] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Background Hyperthermic intraperitoneal chemotherapy (HIPEC) produces unwanted side-effects that are mainly caused by chemotherapeutic drugs in the treatment of gastrointestinal (GI) cancers, and these effects have not been systematically summarized. The aim of this article was to provide a comprehensive overview of the side-effects of HIPEC for GI cancers and propose practical strategies for adverse event management. Methodology PubMed, Web of Science, and the Cochrane Library were systematically searched for side-effects of HIPEC in GI cancers prior to October 20, 2022. A total of 79 articles were included in this review. Results Adverse events, such as enterocutaneous digestive fistulas, GI tract perforation, neutropenia, postoperative bleeding, ventricular tachycardia, hyperglycemia, hypocalcemia, renal impairment, encapsulating peritoneal sclerosis, scrotal ulceration, and sarcopenia were described, and their clinical management was discussed. These side-effects involve the digestive, hematopoietic, circulatory, metabolic, and urinary systems. Effective methods for adverse event management included an expert multidisciplinary team, replacing chemotherapy drugs, using Chinese medicine, and careful preoperative assessments. Conclusion The side-effects of HIPEC are frequent and can be minimized by several effective methods. This study proposes practical strategies for adverse event management of HIPEC to assist physicians in choosing the optimal treatment method.
Collapse
Affiliation(s)
- Jiyun Hu
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenxing Wang
- Department of Hepatobiliary Surgery, Haikou People’s Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Xinrun Wang
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shucai Xie
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Hepatobiliary Surgery, Haikou People’s Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| |
Collapse
|
5
|
Bazarbashi S, Badran A, Gad AM, Aljubran A, Alzahrani A, Alshibani A, Alrakaf R, Elhassan T, Alsuhaibani A, Elshenawy MA. Combined Prophylactic Hyperthermic Intraperitoneal Chemotherapy and Intraoperative Radiotherapy for Localized Gastroesophageal Junction and Gastric Cancer: A Comparative Nonrandomized Study. Ann Surg Oncol 2023; 30:426-432. [PMID: 36042103 DOI: 10.1245/s10434-022-12467-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/09/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The peritoneum frequently is the only recurrence site after radical resection of gastric cancer. Data suggest that hyperthermic intraperitoneal chemotherapy (HIPEC) and intraoperative radiotherapy (IORT) reduce peritoneal recurrence and possibly improve survival for patients with resected gastric and serosal involvement. This study aimed to evaluate the efficacy of combining prophylactic HIPEC and IORT after radical resection of localized gastric cancer. METHODS In this retrospective study, the medical records of adult patients with histologically proven gastric/gastroesophageal adenocarcinoma who underwent radical resection with curative intent were evaluated for recurrence and survival according to whether they received prophylactic HIPEC and IORT. RESULTS The eligibility criteria were met by 58 patients, 33 of whom underwent prophylactic HIPEC and IORT after radical surgery. Overall, 91% the HIPEC/IORT group and 72% of the surgery-only group had ≤pT3 disease. The median follow-up period was 26.6 months for the HIPEC/IORT group and 50.6 months for the surgery group. Locoregional recurrence occurred for six patients (18.1%) in the HIPEC/IORT group and five patients (20%) in the surgery-only group, with peritoneal metastasis (PM) occurring in respectively three (9%) and six (24%) patients. The median recurrence-free survival (RFS) duration was 23.2 months (95% confidence interval [CI] 6.5-39.9 months) for the HIPEC/IORT group versus 24.8 months (95% CI 0.0-51.1 months) for the surgery-only group (p = 0.88), and the corresponding 5-year overall survival (OS) estimates were 69% and 58%. CONCLUSION Prophylactic HIPEC and IORT after radical surgery for localized gastric or gastroesophageal cancer did not improve RFS or OS for an unselected group of patients at risk for peritoneal recurrence.
Collapse
Affiliation(s)
- Shouki Bazarbashi
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| | - Ahmed Badran
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Mostafa Gad
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ali Aljubran
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ahmed Alzahrani
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Aisha Alshibani
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Reem Alrakaf
- Surgical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Tusneem Elhassan
- Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdullah Alsuhaibani
- Radiation Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mahmoud A Elshenawy
- Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| |
Collapse
|
6
|
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: 17] [Impact Index Per Article: 8.5] [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
|
7
|
Choi AY, Singh A, Wang D, Pittala K, Hoang CD. Current State of Pleural-Directed Adjuncts Against Malignant Pleural Mesothelioma. Front Oncol 2022; 12:886430. [PMID: 35586499 PMCID: PMC9108281 DOI: 10.3389/fonc.2022.886430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Multimodality therapy including surgical resection is the current paradigm in treating malignant pleural mesothelioma (MPM), a thoracic surface cancer without cure. The main limitation of all surgical approaches is the lack of long-term durability because macroscopic complete resection (R1 resection) commonly predisposes to locoregional relapse. Over the years, there have been many studies that describe various intrapleural strategies that aim to extend the effect of surgical resection. The majority of these approaches are intraoperative adjuvants. Broadly, there are three therapeutic classes that employ diverse agents. The most common, widely used group of adjuvants are comprised of direct therapeutics such as intracavitary chemotherapy (± hyperthermia). By comparison, the least commonly employed intrathoracic adjuvant is the class comprised of drug-device combinations like photodynamic therapy (PDT). But the most rapidly evolving (new) class with much potential for improved efficacy are therapeutics delivered by specialized drug vehicles such as a fibrin gel containing cisplatin. This review provides an updated perspective on pleural-directed adjuncts in the management of MPM as well as highlighting the most promising near-term technology breakthroughs.
Collapse
|
8
|
Moukarzel LA, Ferrando L, Dopeso H, Stylianou A, Basili T, Pareja F, Da Cruz Paula A, Zoppoli G, Abu-Rustum NR, Reis-Filho JS, Long Roche K, Tew WP, Chi DS, Sonoda Y, Zamarin D, Aghajanian C, O'Cearbhaill RE, Zivanovic O, Weigelt B. Hyperthermic intraperitoneal chemotherapy (HIPEC) with carboplatin induces distinct transcriptomic changes in ovarian tumor and normal tissues. Gynecol Oncol 2022; 165:239-247. [PMID: 35292180 PMCID: PMC9064951 DOI: 10.1016/j.ygyno.2022.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To determine the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) with carboplatin on the transcriptomic profiles of normal and ovarian cancer (OC) tissues. METHODS Normal and tumor samples from four OCs were prospectively collected pre- and immediately post-HIPEC treatment and subjected to RNA-sequencing. Differential gene expression, gene ontology enrichment and pathway analyses were performed. Heat shock protein and immune-response protein expression was assessed using protein arrays and western blotting. RESULTS RNA-sequencing revealed 4231 and 322 genes significantly differentially expressed between pre- and post-treatment normal and OC tissues, respectively (both adjusted p-value <0.05). Gene enrichment analyses demonstrated that the most significantly upregulated genes in normal tissues played a role in immune as well as heat shock response (both adjusted p < 0.001). In contrast, HIPEC induced an increased expression of primarily heat shock response and protein folding-related genes in tumor tissues (both adjusted p < 0.001). HIPEC-induced heat shock protein (HSP) expression changes, including in HSP90, HSP40, HSP60, and HSP70, were also observed at the protein level in both normal and tumor tissues. CONCLUSIONS HIPEC with carboplatin resulted in an upregulation of heat shock-related genes in both normal and tumor tissue, with an additional immune response gene induction in normal and protein folding in tumor tissue. The findings of our exploratory study provide evidence to suggest that HIPEC administration may suffice to induce gene expression changes in residual tumor cells and raises a biological basis for the consideration of combinatorial treatments with HSP inhibitors.
Collapse
Affiliation(s)
- Lea A Moukarzel
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Lorenzo Ferrando
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Higinio Dopeso
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Anthe Stylianou
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Thais Basili
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Arnaud Da Cruz Paula
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Gabriele Zoppoli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Nadeem R Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Kara Long Roche
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - William P Tew
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| | - Dennis S Chi
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Yukio Sonoda
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| | - Roisin E O'Cearbhaill
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America; National University of Ireland, Galway, Ireland
| | - Oliver Zivanovic
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| |
Collapse
|
9
|
Bhatt A, Glehen O. Hyperthermic Intraperitoneal Chemotherapy in the Treatment Armamentarium of Epithelial Ovarian Cancer: Time to End the Dichotomy. Visc Med 2022; 38:109-119. [PMID: 35614893 PMCID: PMC9082174 DOI: 10.1159/000521239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/29/2021] [Indexed: 11/19/2022] Open
Abstract
<b><i>Background:</i></b> Advanced epithelial ovarian cancer (EOC) is an incurable disease with over 75% of the patients developing recurrence in the peritoneum. Hyperthermic intraperitoneal chemotherapy (HIPEC) is a promising treatment option for both first-line therapy and treatment of recurrence. In this article, we review the rationale and current evidence for performing HIPEC and the role of HIPEC in the light of targeted systemic therapies. <b><i>Summary:</i></b> There are few randomized trials and several retrospective studies on the role of HIPEC in the management of EOC. A 12-month-overall survival (OS) benefit of the addition of HIPEC to interval cytoreductive surgery (CRS) was demonstrated in 1 randomized trial following which HIPEC has been included as a treatment option for this indication in several national/international guidelines. One retrospective propensity score-matched analysis showed a 16-month OS benefit of adding HIPEC to primary CRS. One randomized trial showed no benefit of the addition of carboplatin HIPEC to secondary CRS over secondary CRS alone. For patients undergoing primary CRS and secondary CRS for recurrence, the results of ongoing randomized trials are needed to define the role of HIPEC in these situations. All clinical trials have shown that the morbidity of HIPEC performed after CRS is acceptable. Along with the emergence of HIPEC as a promising surgical therapy, targeted therapies like bevacizumab and poly adenosine diphosphate-ribose polymerase inhibitors have been developed that have shown a survival benefit in selected patients. In principle, HIPEC and targeted therapies work in different ways and it is plausible to assume that their benefit could be additive, and their combination should be evaluated in clinical trials. The impact of prognostic factors like the disease extent, pathological response to systemic chemotherapy (SC), the histological subtype and molecular profile on the benefit of HIPEC, and targeted therapies has not been evaluated in clinical trials. <b><i>Key Messages:</i></b> HIPEC is an important therapeutic strategy in the treatment of EOC. While its role in patients undergoing interval CRS has been established, the results of ongoing randomized trials are needed to define its benefit at other time points. The morbidity of HIPEC in addition to CRS is acceptable. More research is needed to define subgroups that benefit most from HIPEC based on the extent of disease, response to SC, histology, and molecular profile. The combination of HIPEC and maintenance therapies should be evaluated in well-designed randomized clinical trials that evaluate not just the survival benefit and morbidity but also the cost-effectiveness of each therapy.
Collapse
Affiliation(s)
- Aditi Bhatt
- Department of Surgical Oncology, Zydus Hospital, Ahmedabad, India
- *Aditi Bhatt,
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-Sud, Lyon, France
| |
Collapse
|
10
|
Wu CC, Hsu YT, Chang CL. Hyperthermic intraperitoneal chemotherapy enhances antitumor effects on ovarian cancer through immune-mediated cancer stem cell targeting. Int J Hyperthermia 2021; 38:1013-1022. [PMID: 34192990 DOI: 10.1080/02656736.2021.1945688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
PURPOSE We aimed to determine the effects and possible mechanisms of hyperthermic intraperitoneal chemotherapy (HIPEC) in targeting ovarian cancer stem-like cells (CSCs). METHODS Murine ovarian cancer cell lines presenting CSC surface markers were grown intraperitoneally in both immunocompetent and immunodeficient mice, which were then treated by intraperitoneal hyperthermia with the chemotherapeutic agents: paclitaxel and cisplatin. Tumor growth was measured by non-invasive luminescent imaging. Intraperitoneal immune cells, such as CD4+, CD8+ T cells, macrophages, and dendritic cells, were evaluated through flow cytometry analysis. RESULTS Combined hyperthermia and chemotherapy exhibited an efficient therapeutic effect in the immunocompetent mice. However, a similar effect was not observed in the immunodeficient mice. Intraperitoneal hyperthermia increased the number of Intraperitoneal macrophages and dendritic cells that were lost due to chemotherapy. Compared with ovarian cancer bulk cells, CSCs were more susceptible to phagocytosis by macrophages. CONCLUSION We demonstrated that the superior therapeutic efficacy and reduced proportion of CSCs associated with intraperitoneal hyperthermic chemotherapy were immune-related. Hyperthermia recruits the phagocytes that target surviving CSCs after chemotherapy. These results provide a novel mechanism for the efficacy of HIPEC in treating ovarian cancer.
Collapse
Affiliation(s)
- Chao-Chih Wu
- Departmental of Medical Research, MacKay Memorial Hospital, Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Yun-Ting Hsu
- Departmental of Medical Research, MacKay Memorial Hospital, Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Chih-Long Chang
- Departmental of Medical Research, MacKay Memorial Hospital, Taipei City, Taiwan.,Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei City, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
11
|
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: 20] [Impact Index Per Article: 6.7] [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
|
12
|
Intraperitoneal Chemotherapy for Peritoneal Metastases: Technical Innovations, Preclinical and Clinical Advances and Future Perspectives. BIOLOGY 2021; 10:biology10030225. [PMID: 33804167 PMCID: PMC8001167 DOI: 10.3390/biology10030225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
(1) Background: Tumors of the peritoneal serosa are called peritoneal carcinosis. Their origin may be primary by primitive involvement of the peritoneum (peritoneal pseudomyxoma, peritoneal mesothelioma, etc.). This damage to the peritoneum can also be a consequence of the dissipation of cancers-in particular, digestive (stomach, pancreas, colorectal, appendix) and gynecological (ovaries) ones in the form of metastases. The aim of the treatment is a maximal reduction of the macroscopic disease called "cytoreduction" in combination with hyperthermic intra-abdominal chemotherapy to treat residual microscopic lesions. (2) Methods: In this narrative review, we fundamentally synthetize the evolution of this process over time and its impact on clinical applications. (3) Results: Over the last past decade, different evolutions concerning both delivery modes and conditions concerning hyperthermic intra-abdominal chemotherapy have been realized. (4) Conclusion: The final objective of these evolutions is the improvement of the global and recurrence-free survival of primary and secondary malignant peritoneal pathologies. However, more large randomized controlled trials are needed to demonstrate the efficacy of such treatments with the help of molecular biology and genetics.
Collapse
|
13
|
Parray A, Gupta V, Chaudhari VA, Shrikhande SV, Bhandare MS. Role of intraperitoneal chemotherapy in gastric cancer. SURGERY IN PRACTICE AND SCIENCE 2021. [DOI: 10.1016/j.sipas.2020.100025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
14
|
Ha HI, Lim MC. How do we perform hyperthermic intraperitoneal chemotherapy in ovarian cancer? -a narrative review. Gland Surg 2021; 10:1235-1243. [PMID: 33842270 DOI: 10.21037/gs-20-425] [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: 11/06/2022]
Abstract
Ovarian malignancy is a leading cause of death caused by gynecologic cancer worldwide because it is mainly found in the advanced stage and recurs in most patients even after cytoreductive surgery and intravenous (IV) chemotherapy. Prevention of recurrence of primary disease and treatment of recurrent ovarian cancer are still remained as major interest and lots of researchers investigate novel treatment to find optimal method. Even though intraperitoneal (IP) chemotherapy turns out to increase the overall survival, it is not widely used because of adverse event. As an alternative treatment for IP chemotherapy, hyperthermic intraperitoneal chemotherapy (HIPEC) is emerging a new way. Thanks to much research and use in other cancer species, such as the colorectal cancer cytoreductive surgery followed by HIPEC is becoming a promising treatment. However, randomized controlled trials and unbiased data in ovarian cancer patients are still needed for the establishment of therapy. Moreover, among the current situation in which treatments such as bevacizumab or PARP inhibitor have been found to be effective and have been widely used, it may be necessary to establish the role in the combination of HIPEC. This article is a comprehensive review of the HIPEC in ovarian cancer to introduce techniques, treatment results, and clinical trials of HIPEC.
Collapse
Affiliation(s)
- Hyeong In Ha
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Myong Cheol Lim
- Research Institute and Hospital, National Cancer Center, Goyang, Korea
| |
Collapse
|
15
|
Yin Z, Wei M, Xie S, Zhou S, Zhang B, Gao P, Wu T, Qiao Q, Wang N, He X. Laparoscopic distal gastrectomy and hyperthermic intraperitoneal chemotherapy in the treatment of advanced gastric cancer: a retrospective case-matched study on perioperative outcomes. J Gastrointest Oncol 2021; 12:133-141. [PMID: 33708431 DOI: 10.21037/jgo-21-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background This study aimed to assess the safety and efficacy of laparoscopic distal gastrectomy (LDG) with intraoperative hyperthermic intraperitoneal chemotherapy (HIPEC) for advanced gastric cancer (AGC). Methods In this case-matched study, we retrospectively reviewed the database of 223 patients with AGC who underwent LDG in Tangdu Hospital from April 2016 to February 2019. Among all participants, 177 patients underwent LDG alone and 46 underwent LDG with HIPEC. We matched total of 138 (1:2) patients from the LDG + HIPEC group (n 46) and the LDG group (n 92) for gender, age, date of operation, and tumor-node-metastasis (TNM) stage of tumor. Results There was no significant difference in the Clavien-Dindo classification of complications between LDG alone and LDG + HIPEC patients. Further analysis showed the morbidity of gastroparesis to be significantly increased in LDG + HIPEC patients. At the same time, we found that the operation time, the time to 1st flatus, and hospital stay were longer in LDG + HIPEC patients and the incidence of abdominal recurrence 2 years after operation was significantly higher in the LDG group than the LDG + HIPEC group. Conclusions The combination of LDG with intraoperative HIPEC is a safe and feasible method for AGC and HIPEC will limit the recovery of gastrointestinal functions. In addition, during the follow-up of our study, although there was no statistical difference between the two groups in abdominal recurrence at 2 years after surgery, a decreasing trend of abdominal recurrence in LDG + HIPEC patients could be seen in comparison to LDG patients.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Mingguang Wei
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Shuang Xie
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Shuai Zhou
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Bo Zhang
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Peng Gao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Tao Wu
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Nan Wang
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
16
|
Survival after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for colorectal peritoneal metastases: A systematic review and discussion of latest controversies. Surgeon 2020; 19:310-320. [PMID: 33023847 DOI: 10.1016/j.surge.2020.08.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/14/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Peritoneal metastases confer the worst survival amongst all sites of metastatic colorectal cancer. The adoption of cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) has become an option for patients with isolated colorectal peritoneal metastases (CRPM). The aim of this study was to evaluate the outcomes following CRS and HIPEC for CRPM from published high volume cohort studies and to highlight the latest controversies and future directions of CRPM treatment. MATERIALS AND METHODS A systematic review was performed on published studies on the treatment outcomes of CRS and HIPEC for colorectal peritoneal metastases. RESULTS Twenty studies met the inclusion criteria for the systematic review. The median survival for all patients ranged from 14.6 to 60.1 months. The 5-year overall survival ranged from 23.4% to 52%. For patients with complete cytoreduction, the median survival was 25 to 49 months. Major morbidity and mortality ranged from 15.1% to 47.2% and 0% to 4.5%, respectively. CONCLUSION CRS and HIPEC for the treatment of CRPM is safe and current evidence suggests it improves both median and disease-free survival. However, the efficacy of intraperitoneal chemotherapy, in particular oxaliplatin, has recently come under scrutiny. Accordingly, higher quality evidence is urgently required to contribute to multidisciplinary and international consensus on CRPM treatment strategies.
Collapse
|
17
|
Chambers LM, Costales AB, Crean-Tate K, Kuznicki M, Morton M, Horowitz M, Jagielo T, Rose PG, Michener C, Vargas R, Debernardo R. A guide to establishing a hyperthermic intraperitoneal chemotherapy program in gynecologic oncology. Gynecol Oncol 2020; 158:794-802. [DOI: 10.1016/j.ygyno.2020.06.487] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
|
18
|
Forsythe SD, Sasikumar S, Moaven O, Sivakumar H, Shen P, Levine EA, Soker S, Skardal A, Votanopoulos KI. Personalized Identification of Optimal HIPEC Perfusion Protocol in Patient-Derived Tumor Organoid Platform. Ann Surg Oncol 2020; 27:4950-4960. [PMID: 32632882 DOI: 10.1245/s10434-020-08790-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/18/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Chemotherapy dosing duration and perfusion temperature vary significantly in HIPEC protocols. This study investigates patient-derived tumor organoids as a platform to identify the most efficacious perfusion protocol in a personalized approach. PATIENTS AND METHODS Peritoneal tumor tissue from 15 appendiceal and 8 colon cancer patients who underwent CRS/HIPEC were used for personalized organoid development. Organoids were perfused in parallel at 37 and 42 °C with low- and high-dose oxaliplatin (200 mg/m2 over 2 h vs. 460 mg/m2 over 30 min) and MMC (40 mg/3L over 2 h). Viability assays were performed and pooled for statistical analysis. RESULTS An adequate organoid number was generated for 75% (6/8) of colon and 73% (11/15) of appendiceal patients. All 42 °C treatments displayed lower viability than 37 °C treatments. On pooled analysis, MMC and 200 mg/m2 oxaliplatin displayed no treatment difference for either appendiceal or colon organoids (19% vs. 25%, p = 0.22 and 27% vs. 31%, p = 0.55, respectively), whereas heated MMC was superior to 460 mg/m2 oxaliplatin in both primaries (19% vs. 54%, p < 0.001 and 27% vs. 53%, p = 0.002, respectively). In both appendiceal and colon tumor organoids, heated 200 mg/m2 oxaliplatin displayed increased cytotoxicity as compared with 460 mg/m2 oxaliplatin (25% vs. 54%, p < 0.001 and 31% vs. 53%, p = 0.008, respectively). CONCLUSIONS Organoids treated with MMC or 200 mg/m2 heated oxaliplatin for 2 h displayed increased susceptibility in comparison with 30-min 460 mg/m2 oxaliplatin. Optimal perfusion protocol varies among patients, and organoid technology may offer a platform for tailoring HIPEC conditions to the individual patient level.
Collapse
Affiliation(s)
- Steven D Forsythe
- Wake Forest Organoid Research Center (WFORCE), Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shyama Sasikumar
- Wake Forest Organoid Research Center (WFORCE), Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Omeed Moaven
- Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA.,Department of Surgery - Surgical Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Hemamylammal Sivakumar
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.,The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Perry Shen
- Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA.,Department of Surgery - Surgical Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Edward A Levine
- Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA.,Department of Surgery - Surgical Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Shay Soker
- Wake Forest Organoid Research Center (WFORCE), Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.,The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Konstantinos I Votanopoulos
- Wake Forest Organoid Research Center (WFORCE), Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Comprehensive Cancer Center at Wake Forest Baptist Medical, Winston-Salem, NC, USA. .,Department of Surgery - Surgical Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
19
|
Ye J, Chen L, Zuo J, Peng J, Chen C, Cai S, Song W, He Y, Yuan Y. A precise temperature control during hyperthermic intraperitoneal chemotherapy promises an early return of bowel function. Cancer Biol Ther 2020; 21:726-732. [PMID: 32644887 PMCID: PMC7515524 DOI: 10.1080/15384047.2020.1775444] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction The hyperthermic intraperitoneal chemotherapy (HIPEC) has been widely applied in clinical practice for peritoneal carcinomatosis (PC). The temperature is one of the important elements affecting the efficacy of HIPEC, and it can become fluctuant by several factors. This study is aimed to explore the role of a stable perfusion temperature in promoting bowel recovery of PC patients due to gastrointestinal malignancies. Methods Between January 2012 and July 2013, 59 PC patients undergoing cytoreductive surgery and three-cycle HIPEC were included. Patients having stable perfusion temperature for all cycles were assigned into the study group, with the rest having unstable temperature into the control group. Time of flatus and defecation passage and initiation time of enteral nutrition were compared between both groups to detect the significance in bowel function recovery, with visual analogue scale (VAS) pain intensity and overall survival (OS) compared meanwhile. Results In sum, 33 (55.9%) patients obtained stable temperature during HIPEC, and the rest of 26 (44.1%) developed fluctuant perfusion temperature. Average time of flatus (2.3 ± 1.2 vs 3.9 ± 2.2 days, P =.002), defecation passage (5.2 ± 2.1 vs 7.1 ± 2.9 days, P =.004) and enteral nutrition initiation (4.3 ± 1.5 vs 6.7 ± 2.3 days, P <.001) were much shorter in the study group than the control group. Additionally, the VAS score (4.5 ± 2.3 vs 6.3 ± 1.3, P <.001) and 5-year OS rate (17.8% vs 11.1%, P=.135) were both improved, with significance observed in postoperative pain control. Conclusions During HIPEC, a precise temperature control could promise an early recovery of bowel function and reduce postoperative pain, without survival significance found based on the current cohort.
Collapse
Affiliation(s)
- Jinning Ye
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Liuhua Chen
- Department of Biliary-Pancreatic Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Jidong Zuo
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Jianjun Peng
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Chuangqi Chen
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Shirong Cai
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Wu Song
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Yulong He
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Yujie Yuan
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| |
Collapse
|
20
|
de Bree E, Michelakis D. An overview and update of hyperthermic intraperitoneal chemotherapy in ovarian cancer. Expert Opin Pharmacother 2020; 21:1479-1492. [PMID: 32486865 DOI: 10.1080/14656566.2020.1766024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Despite, the strong rationale and evidence of the benefit of postoperative intraperitoneal chemotherapy in advanced ovarian cancer, it has not been widely adopted, mainly due to its high morbidity and logistical difficulties. Intraoperative hyperthermic intraperitoneal chemotherapy (HIPEC) is a more tolerable and technically feasible method of intraperitoneal chemotherapy, whereas other potential advantages include homogenous drug distribution, application before tumor regrowth and combination with hyperthermia, which is directly cytotoxic and enhances the efficacy of many drugs. AREAS COVERED In this review, the authors explain the rationale and indications for cytoreductive surgery (CRS) and HIPEC in advanced ovarian cancer. Data of major clinical studies, meta-analyses, and recent randomized trials are discussed. EXPERT OPINION After many encouraging clinical studies and meta-analyses, a recent randomized study demonstrated survival benefit for HIPEC during interval CRS in primary ovarian cancer, without increased morbidity, whereas another implied its benefit in recurrent ovarian cancer. Results of recently completed and numerous ongoing randomized studies will further determine the benefit of HIPEC in ovarian cancer at different time points. Patient selection and appraisal of the best protocols are crucial. The field of gynecological oncology will most likely evolve to include HIPEC eventually as a routine treatment for ovarian cancer.
Collapse
Affiliation(s)
- Eelco de Bree
- Department of Surgical Oncology, Medical School of Crete University Hospital , Heraklion, Greece
| | - Dimosthenis Michelakis
- Department of Surgical Oncology, Medical School of Crete University Hospital , Heraklion, Greece
| |
Collapse
|
21
|
Study protocol of a multicenter phase III randomized controlled trial investigating the efficiency of the combination of neoadjuvant chemotherapy (NAC) and neoadjuvant laparoscopic intraperitoneal hyperthermic chemotherapy (NLHIPEC) followed by R0 gastrectomy with intraoperative HIPEC for advanced gastric cancer (AGC): dragon II trial. BMC Cancer 2020; 20:224. [PMID: 32183736 PMCID: PMC7077163 DOI: 10.1186/s12885-020-6701-2] [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: 11/19/2019] [Accepted: 02/28/2020] [Indexed: 01/14/2023] Open
Abstract
Background Even though treatment modalities such as adjuvant systemic radio-chemotherapy and neoadjuvant chemotherapy (NAC) have individually have improved overall survival (OS) and progression-free survival (PFS) rates in advanced Gastric Cancer (AGC), the peritoneum still presides as a common site of treatment failure and disease recurrence. The role of hyperthermic intraperitoneal chemotherapy (HIPEC) has been acknowledged as prophylaxis for peritoneal carcinomatosis (PC) in AGC patients and in this study, we aim at investigating the safety and efficacy of the combination of neoadjuvant laparoscopic HIPEC (NLHIPEC) with NAC in the neoadjuvant phase followed by surgery of curative intent with intraoperative HIPEC followed by adjuvant chemotherapy (AC). Methods In this multicenter Phase III randomized controlled trial, 326 patients will be randomly separated into 2 groups into a 1:1 ratio after laparoscopic exploration. The experiment arm will receive the proposed comprehensive Dragon II regimen while the control group will undergo standard R0 D2 followed by 8 cycles of AC with oxaliplatin with S-1 (SOX) regimen. The Dragon II regimen comprises of 1 cycle of NLHIPEC for 60mins at 43 ± 0.5 °C with 80 mg/m2 of Paclitaxel followed by 3 cycles of NAC with SOX regimen and after assessment, standard R0 D2 gastrectomy with intraoperative HIPEC followed by 5 cycles of SOX regimen chemotherapy. The end-points for the study are 5 year PFS, 5 year OS, peritoneal metastasis rate (PMR) and morbidity rate. Discussion This study is one of the first to combine NLHIPEC with NAC in the preoperative phase which is speculated to provide local management of occult peritoneal carcinomatosis or peritoneal free cancer cells while NAC will promote tumor downsizing and down-staging. The addition of the intraoperative HIPEC is speculated to manage dissemination due to surgical trauma. Where the roles of intraoperative HIPEC and NAC have individually been investigated, this study provides innovative insight on a more comprehensive approach to management of AGC at high risk of peritoneal recurrence. It is expected that the combination of NLHIPEC with NAC and HIPEC will increase PFS by 15% and decrease PMR after gastrectomy of curative intent. Trial registration World Health Organization Clinical Trials - International Registry Platform (WHO-ICTRP) with Registration ID ChiCTR1900024552, Registered Prospectively on the 16th July, 2019.
Collapse
|
22
|
Emerging hyperthermia applications for pediatric oncology. Adv Drug Deliv Rev 2020; 163-164:157-167. [PMID: 33203538 DOI: 10.1016/j.addr.2020.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022]
Abstract
Local application of hyperthermia has a myriad of effects on the tumor microenvironment as well as the host's immune system. Ablative hyperthermia (typically > 55 °C) has been used both as monotherapy and adjuvant therapy, while mild hyperthermia treatment (39-45 °C) demonstrated efficacy as an adjuvant therapy through enhancement of both chemotherapy and radiation therapy. Clinical integration of hyperthermia has especially great potential in pediatric oncology, where current chemotherapy regimens have reached maximum tolerability and the young age of patients implies significant risks of late effects related to therapy. Furthermore, activation of both local and systemic immune response by hyperthermia suggests that hyperthermia treatments could be used to enhance the anticancer effects of immunotherapy. This review summarizes the state of current applications of hyperthermia in pediatric oncology and discusses the use of hyperthermia in the context of other available treatments and promising pre-clinical research.
Collapse
|
23
|
Cytoreductive Surgery With Hyperthermic Intraperitoneal Chemotherapy, Part I: Introduction and Indications. AORN J 2019; 110:479-499. [DOI: 10.1002/aorn.12842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
24
|
Yarema R, Volodko N, Fetsych T, Оhorchak M, Petronchak O, Mylyan Y, Makukh H. Cytoreductive surgery and HIPEC (hyperthermic intraperitoneal chemotherapy) in combined treatment of ovarian cancer: time for the beginning of personalized therapy? CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2019; 32:154-159. [DOI: 10.2478/cipms-2019-0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract
Background and objectives. During the two past decades, a new therapeutic approach to ovarian cancer (OC) has been developed. This combines cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). However, almost no data exist regarding the utility of biomarkers of morphological heterogeneity as prognostic factors in such patients.
Methods. A retrospective study of the effectiveness of CRS and HIPEC was carried out in 59 patients with ovarian cancer. Biomarkers of morphological heterogeneity of OC were studied as prognostic factors: OC pathogenic types (based on the identification of р53 mutated gene protein expression) and homologous recombination deficit (basing on the identification of BRCA 1 gene expression status).
Results. The survival of patients reliably differed with the division into two pathogenetic OC types established by immunohistochemistry: the median disease-free survival of type I OC patients was 14±1.7 months, type ІІ – 8±1.6 months (р = 0.007); the median overall survival of type I OC patients was 23.5±6.7 months, type ІІ – 12±1.9 months (р = 0.017). The median overall survival of patients with the somatic mutation of BRCA 1 gene and complete cytoreduction was 22±4.8 months, and without the somatic mutation of BRCA 1 gene – 12±3.3 months (р = 0.047).
Conclusions. These data demonstrate that identification of the pathogenetic type of OC and BRCA 1 status may be useful for the personalized therapy of ovarian cancer patients treated with CRS/HIPEC.
Collapse
Affiliation(s)
- Roman Yarema
- Danylo Halytsky Lviv National Medical University , Department of Oncology and Medical Radiology , Lviv , Ukraine
| | - Nataliya Volodko
- Danylo Halytsky Lviv National Medical University , Department of Oncology and Medical Radiology , Lviv , Ukraine
| | - Taras Fetsych
- Danylo Halytsky Lviv National Medical University , Department of Oncology and Medical Radiology , Lviv , Ukraine
| | - Myron Оhorchak
- Lviv State Oncological Regional Treatment and Diagnostic Center , Lviv , Ukraine
| | - Orest Petronchak
- Lviv State Oncological Regional Treatment and Diagnostic Center , Lviv , Ukraine
| | - Yuriy Mylyan
- Lviv State Oncological Regional Treatment and Diagnostic Center , Lviv , Ukraine
| | - Halyna Makukh
- SI “Institute of Hereditary Pathology Ukrainian National Academy of Medical Sciences” , Lviv , Ukraine
| |
Collapse
|
25
|
|
26
|
Sugarbaker PH. Prevention and Treatment of Peritoneal Metastases: a Comprehensive Review. Indian J Surg Oncol 2019; 10:3-23. [PMID: 30948866 PMCID: PMC6414583 DOI: 10.1007/s13193-018-0856-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/04/2018] [Indexed: 02/06/2023] Open
Abstract
Peritoneal metastases may occur from a majority of cancers that occur within the abdomen or pelvis. When cancer spread to the peritoneal surfaces is documented, a decision regarding palliation versus an aggressive approach using cytoreductive surgery (CRS) and hyperthermic perioperative intraperitoneal chemotherapy (HIPEC) must be made. This decision is dependent on a well-defined group of prognostic indicators. In addition to treatment, prevention of peritoneal metastases may be an option. The clinical and pathologic features of a primary cancer can be used to select perioperative treatments that may prevent cancer cells within the abdomen and pelvis from progressing to established peritoneal metastases. In some clinical situations with appendiceal and colorectal cancers, the clinical or histopathologic features may indicate that second-look surgery plus perioperative chemotherapy should occur. Peritoneal metastases should always be considered by the multidisciplinary team for treatment or prevention.
Collapse
Affiliation(s)
- Paul H. Sugarbaker
- Center for Gastrointestinal Malignancies, MedStar Washington Hospital Center, 106 Irving St., NW, Suite 3900, Washington, DC 20010 USA
| |
Collapse
|
27
|
González-Moreno S, Ortega-Pérez G, Alonso-Casado O, Galipienzo-García J, Linero-Noguera MJ, Salvatierra-Díaz D. Techniques and Safety Issues for Intraperitoneal Chemotherapy. Surg Oncol Clin N Am 2018; 27:495-506. [DOI: 10.1016/j.soc.2018.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
28
|
Mas-Fuster MI, Ramon-Lopez A, Lacueva FJ, Arroyo A, Más-Serrano P, Nalda-Molina R. Population pharmacokinetics of oxaliplatin after intraperitoneal administration with hyperthermia in Wistar rats. Eur J Pharm Sci 2018; 119:22-30. [PMID: 29626594 DOI: 10.1016/j.ejps.2018.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/20/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The evaluation of the efficacy and toxicity of hyperthermic intraoperative peritoneal chemotherapy presents some difficulties, due in part to the lack of information about the pharmacokinetic behavior of the drugs administered in this procedure. The aim of this study was to characterize the population pharmacokinetics of hyperthermic intraoperative peritoneal oxaliplatin in Wistar rats and to evaluate the effect of treatment-related covariates dose, instillation time and temperature on the pharmacokinetic parameters. METHODS Oxaliplatin peritoneal and plasma concentrations from 37 rats treated by either intravenous or intraperitoneal oxaliplatin administrations under different instillation times, temperatures and doses were analyzed according to a population pharmacokinetic approach using the software NONMEM V7.3®. RESULTS Intraperitoneal (n = 115) and plasma (n = 263) concentrations were successfully described according to a two-compartment model with first order absorption. No significant effect of dose, temperature and instillation time on pharmacokinetic parameters was found. However, an abrupt decrease in the elimination process was observed, reflected in the structural pharmacokinetic model through a modification in clearance. The typical parameters values and the interindividual variability (CV %) in clearance, central and peripheral volume of distribution were 3.25 mL/min (39.1%), 53.6 mL (37.8%) and 54.1 mL (77.3%), respectively. Clearance decreased to 0.151 mL/min (39.1%) when the instillation was still ongoing, at 31.4 min. One of the possible reasons behind the clearance decrease would be an alteration of renal function due to surgery and/or hyperthermia. CONCLUSIONS This study described the deterioration of the drug elimination process due to the procedure, and estimated the time at which this deterioration is most likely to occur. In addition, dose, instillation time and temperature had no influence in the PK parameters.
Collapse
Affiliation(s)
- M I Mas-Fuster
- Division of Pharmacy and Pharmaceutics, Department of Engineering, School of Pharmacy, Miguel Hernández University, San Juan de Alicante, Alicante, Spain
| | - A Ramon-Lopez
- Division of Pharmacy and Pharmaceutics, Department of Engineering, School of Pharmacy, Miguel Hernández University, San Juan de Alicante, Alicante, Spain; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante, Spain.
| | - F J Lacueva
- Department of Pathology and Surgery, School of Medicine, Miguel Hernández University, San Juan de Alicante, Alicante, Spain.
| | - A Arroyo
- Department of Pathology and Surgery, School of Medicine, Miguel Hernández University, San Juan de Alicante, Alicante, Spain.
| | - P Más-Serrano
- Division of Pharmacy and Pharmaceutics, Department of Engineering, School of Pharmacy, Miguel Hernández University, San Juan de Alicante, Alicante, Spain; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante, Spain; Clinical Pharmacokinetics Unit, Pharmacy Department, Hospital General Universitario de Alicante, Alicante, Spain
| | - R Nalda-Molina
- Division of Pharmacy and Pharmaceutics, Department of Engineering, School of Pharmacy, Miguel Hernández University, San Juan de Alicante, Alicante, Spain; Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante, Spain.
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Peritoneal metastases may occur from a majority of cancers that occur within the abdomen or pelvis. When cancer spread to the peritoneal surfaces is documented, a decision regarding palliation vs. an aggressive approach using cytoreductive surgery (CRS) and perioperative intraperitoneal chemotherapy must be made. The perioperative chemotherapy may be hyperthermic intraperitoneal chemotherapy (HIPEC) administered in the operating room or early postoperative intraperitoneal chemotherapy (EPIC) administered in the first 4 or 5 postoperative days. RECENT FINDINGS This decision is dependent on a well-defined group of prognostic indicators. In addition to treatment, the clinical and pathologic features of a primary cancer can be used to select perioperative treatments that may prevent cancer cells within the abdomen and pelvis from progressing to established peritoneal metastases. In some clinical situations with appendiceal and colorectal cancers, the clinical or histopathologic features may indicate that second-look surgery plus perioperative chemotherapy should occur. Peritoneal metastases should always be considered for treatment or prevention.
Collapse
|
30
|
Mehta S, Schwarz L, Spiliotis J, Hsieh MC, Akaishi EH, Goere D, Sugarbaker PH, Baratti D, Quenet F, Bartlett DL, Villeneuve L, Kepenekian V. Is there an oncological interest in the combination of CRS/HIPEC for peritoneal carcinomatosis of HCC? Results of a multicenter international study. Eur J Surg Oncol 2018; 44:1786-1792. [PMID: 29885982 DOI: 10.1016/j.ejso.2018.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/22/2018] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Peritoneal metastasis (PM) of hepatocellular carcinoma (HCC) without distant spread are rare. The related prognosis is poor without standard treatment available. The role of cytoreduction surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) is poorly documented. METHODS An international multicentric cohort was constituted by retrospective analysis of 21 patients undergoing CRS/HIPEC for PM of HCC between 1992 and 2016 from 10 reference centers of PSOGI. Data on clinical features, treatment strategies, and survival outcomes were analyzed. RESULTS The median time interval from the diagnosis of PM to the procedure was 4.5 months. The median peritoneal cancer index was 14. Sixteen patients had complete cytoreduction (CCR0-1). Ten patients had grades 3 to 4 complications. The median duration of follow-up was 52.2 months. The median OS was 46.7 months. The projected 3y-OS and 5y-OS were 88.9 and 49.4% respectively. The median OS for patients with CCR0-1 resection was not reached whereas it was 5.9 months for those with CCR2-3 resection after CRS (p = 0.0005). The median RFS was 26.3 months and projected RFS at 3 years of 36.5 months Three prognostic factors were associated with improved RFS in the univariate analysis: preoperative chemotherapy (p = 0.0156), PCI >15 (p = 0.009), Number of chemotherapy agents used for HIPEC (p = 0.005). CONCLUSION CRS/HIPEC is a safe and effective approach in selected patients with PM of HCC. CRS/HIPEC gives the patient a chance for a good relapse free and overall survival and should be considered as an option.
Collapse
Affiliation(s)
- Sanket Mehta
- Department of Peritoneal Surface Oncology, Saifee Hospital, Mumbai, India
| | - Lilian Schwarz
- Department of Digestive Surgery, Hôpital Charles Nicolle, Rouen University Hospital, Rouen, France.
| | - John Spiliotis
- First Department of Surgical Oncology, Metaxa Cancer Memorial Hospital, Piraeus, Greece
| | - Mao-Chih Hsieh
- Department of General Surgery, Wan-Fang Hospital, Tapei, Taiwan
| | - Eduardo H Akaishi
- Department of Surgical Oncology, Centro de Oncologia Hospital Sirio Libanes, Sao Paulo, Brazil
| | - Diane Goere
- Department of Surgical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Paul H Sugarbaker
- Department of Surgical Oncology, Washington Hospital Center, Washington, United States
| | - Dario Baratti
- Department of Gastrointestinal Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - François Quenet
- Department of Surgical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh Medical Center Shaydyside Hospital, Pittsburgh, United States
| | - Laurent Villeneuve
- Hospices Civils de Lyon, Pôle Information Médicale Evaluation Recherche, Unité de Recherche Clinique, Lyon, France; EMR 3738, Lyon 1 University, Lyon, France
| | - Vahan Kepenekian
- EMR 3738, Lyon 1 University, Lyon, France; Department of Digestive Surgery, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| | | |
Collapse
|
31
|
Dodson RM, Kuncewitch M, Votanopoulos KI, Shen P, Levine EA. Techniques for Cytoreductive Surgery with Hyperthermic Intraperitoneal Chemotherapy. Ann Surg Oncol 2018; 25:2152-2158. [PMID: 29388121 DOI: 10.1245/s10434-018-6336-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 01/26/2023]
Affiliation(s)
- Rebecca M Dodson
- Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA
| | - Michael Kuncewitch
- Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA
| | | | - Perry Shen
- Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA
| | - Edward A Levine
- Surgical Oncology, Wake Forest Baptist Health, Wake Forest University, Winston-Salem, NC, USA.
| |
Collapse
|
32
|
Hornung M, Werner JM, Schlitt HJ. Applications of hyperthermic intraperitoneal chemotherapy for metastatic colorectal cancer. Expert Rev Anticancer Ther 2017; 17:841-850. [PMID: 28715968 DOI: 10.1080/14737140.2017.1357470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) plays a pivotal role in the current treatment of peritoneal carcinomatosis (PC) from colorectal cancer (CRC). Since the first demonstration, benefits for patients and especially an increase in survival have been described. In recent years, feasibility, efficacy and safety of HIPEC have been improved and progress has been made in understanding its oncological mechanism. Areas covered: In this article, leading publications have been reviewed including clinical trials to describe the clinical presentation of PC due to CRC and present recent evidence of the CRS/HIPEC procedure. The surgical approach including evaluation of the extent of PC is described and, in addition, the article reports about different HIPEC techniques as well as several protocols. Furthermore, the development and prognostic benefit of the combination of intraperitoneal and intravenous chemotherapy are outlined. Consideration has been given in particular to patient selection and the use of HIPEC if complete cytoreduction is not feasible. Expert commentary: The CRS/HIPEC procedure represents a curative approach to treat patients with PC from CRC. However, surgical skills and the HIPEC technique still require specialized oncological centers.
Collapse
Affiliation(s)
- Matthias Hornung
- a Department of Surgery , University of Regensburg , Regensburg , Germany
| | - Jens M Werner
- a Department of Surgery , University of Regensburg , Regensburg , Germany
| | - Hans J Schlitt
- a Department of Surgery , University of Regensburg , Regensburg , Germany
| |
Collapse
|
33
|
de Bree E, Michelakis D, Stamatiou D, Romanos J, Zoras O. Pharmacological principles of intraperitoneal and bidirectional chemotherapy. Pleura Peritoneum 2017; 2:47-62. [PMID: 30911633 PMCID: PMC6405033 DOI: 10.1515/pp-2017-0010] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/05/2017] [Indexed: 12/19/2022] Open
Abstract
Intraperitoneal chemotherapy is associated with a significant pharmacokinetic and pharmacodynamic benefit and can, alone or in combination with systemic chemotherapy (bidirectional chemotherapy), be used for treating primary and secondary peritoneal surface malignancies. Due to the peritoneal-plasma barrier, high intraperitoneal drug concentration can be achieved by intraperitoneal chemotherapy, whereas systemic concentration remains low. Bidirectional chemotherapy may provide in addition adequate drug concentrations from the side of the subperitoneal space to the peritoneal tumour nodules. Major pharmacological problems of intraperitoneal chemotherapy are limited tissue penetration and poor homogeneity of drug distribution to the entire seroperitoneal surface. Significant pharmacological determinants of intraperitoneal chemotherapy are choice of drug, drug dosage, solution volume, carrier solution, intra-abdominal pressure, temperature, duration, mode of administration, extent of peritonectomy and interindividual variability. Drugs most commonly applied for intraperitoneal chemotherapy include mitomycin C, cisplatin, carboplatin, oxaliplatin, irinotecan, 5-fluoruracil, gemcitabine, paclitaxel, docetaxel, doxorubicin, premetrexed and melphalan. The drugs and their doses that are used vary widely among centres. While the adequate drug choice for intraperitoneal and bidirectional chemotherapy is essential, randomized clinical trials to determine the most optimal drug or drug combination are lacking, and only eight retrospective comparative clinical studies are available. Further clinical pharmacological studies are required to determine the most effective drug regimen for intraperitoneal and bidirectional chemotherapy in various indications. In the future, reliable drug sensitivity testing and genetic profiling of peritoneal metastases will be needed for enabling patient-specific therapy.
Collapse
Affiliation(s)
- Eelco de Bree
- Department of Surgical Oncology, Medical School of Crete University Hospital, Heraklion, Greece
| | - Dimosthenis Michelakis
- Department of Surgical Oncology, Medical School of Crete University Hospital, Heraklion, Greece
| | - Dimitris Stamatiou
- Department of Surgical Oncology, Medical School of Crete University Hospital, Heraklion, Greece
| | - John Romanos
- Department of Surgical Oncology, Medical School of Crete University Hospital, Heraklion, Greece
| | - Odysseas Zoras
- Department of Surgical Oncology, Medical School of Crete University Hospital, Heraklion, Greece
| |
Collapse
|
34
|
Zhou H, Wu W, Tang X, Zhou J, Shen Y. Effect of hyperthermic intrathoracic chemotherapy (HITHOC) on the malignant pleural effusion: A systematic review and meta-analysis. Medicine (Baltimore) 2017; 96:e5532. [PMID: 28072694 PMCID: PMC5228654 DOI: 10.1097/md.0000000000005532] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Although hyperthermic intraperitoneal chemotherapy (HIPEC) has been widely used to treat malignant ascites or as a preventive strategy for microscopic carcinomatosis following surgical resection of abdominal tumors, application of hyperthermic intrathoracic chemotherapy (HITHOC) in the treatment of malignant pleural effusion is limited. The objective of the current study was to conduct a systematic review and meta-analysis on the application of HITHOC in the palliative treatment of malignant pleural effusion. METHODS After thorough searching of online databases, total 27 articles were included into qualitative systematic review and 5 of them were used to conduct qualitative meta-analysis. RESULTS It was found that most of HITHOC was used in combination of cytoreductive surgery (CRS) including pleurectomy/decortication or after surgical resection of primary tumors, which mainly were lung cancer, thymoma or thymic carcinoma, breast cancer, and ovarian cancer. Patients who received HITHOC had significantly longer median survival length compared to the patients without HITHOC (Hedges g = 0.763, P < 0.001). In addition, HITHOC therapy was favored (Hedges g = 0.848, P < 0.001) in terms of median survival length, tumor-free survival rate, with tumor survival rate or Karnofsky performance status (KPS) scale. CONCLUSION HITHOC is a safe and effective therapy in controlling pleural effusion and increasing patient's survival rate.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Respiration, The First Affiliated Hospital, Zhejiang University, Hangzhou
| | - Wei Wu
- Department of Radiology, Jilin Provincial Tumor Hospital, Changchun, China
| | - Xiaoping Tang
- Department of Respiration, The First Affiliated Hospital, Zhejiang University, Hangzhou
| | - Jianying Zhou
- Department of Respiration, The First Affiliated Hospital, Zhejiang University, Hangzhou
| | - Yihong Shen
- Department of Respiration, The First Affiliated Hospital, Zhejiang University, Hangzhou
| |
Collapse
|
35
|
Ji ZH, Peng KW, Li Y. Intraperitoneal free cancer cells in gastric cancer: pathology of peritoneal carcinomatosis and rationale for intraperitoneal chemotherapy/hyperthermic intraperitoneal chemotherapy in gastric cancer. Transl Gastroenterol Hepatol 2016; 1:69. [PMID: 28138635 DOI: 10.21037/tgh.2016.08.03] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 08/15/2016] [Indexed: 12/19/2022] Open
Abstract
Peritoneal carcinomatosis (PC) is one of the most common causes of death in gastric cancer patients. Intraperitoneal free cancer cells (IFCCs) play a very important role in forming PC, but the administration of intraperitoneal chemotherapy (IPC) and/or hyperthermic intraperitoneal chemotherapy (HIPEC) could be an effective treatment for IFCCs. This review focuses on the origin of IFCCs, the mechanism of PC formatting, the rationale of IPC/HIPEC, and the current clinical trials on IPC/HIPEC to treat advanced gastric cancer patients.
Collapse
Affiliation(s)
- Zhong-He Ji
- Department of Peritoneal Cancer Surgery, Cancer Center of Beijing Shijitan Hospital affiliated to the Capital Medical University, Beijing 100038, China
| | - Kai-Wen Peng
- Department of Peritoneal Cancer Surgery, Cancer Center of Beijing Shijitan Hospital affiliated to the Capital Medical University, Beijing 100038, China
| | - Yan Li
- Department of Peritoneal Cancer Surgery, Cancer Center of Beijing Shijitan Hospital affiliated to the Capital Medical University, Beijing 100038, China
| |
Collapse
|
36
|
Chia CS, Seshadri RA, Kepenekian V, Vaudoyer D, Passot G, Glehen O. Survival outcomes after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis from gastric cancer: a systematic review. Pleura Peritoneum 2016; 1:67-77. [PMID: 30911610 PMCID: PMC6386497 DOI: 10.1515/pp-2016-0010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 05/08/2016] [Indexed: 02/07/2023] Open
Abstract
Background: The current treatment of choice for peritoneal carcinomatosis from gastric cancer is systemic chemotherapy. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) is a new aggressive form of loco-regional treatment that is currently being used in pseudomyxoma peritoneii, peritoneal mesothelioma and peritoneal carcinomatosis from colorectal cancer. It is still under investigation for its use in gastric cancer. Methods: The literature between 1970 and 2016 was surveyed systematically through a review of published studies on the treatment outcomes of CRS and HIPEC for peritoneal carcinomatosis from gastric cancer. Results: Seventeen studies were included in this review. The median survival for all patients ranged from 6.6 to 15.8 months. The 5-years overall survival ranged from 6 to 31%. For patients with complete cytoreduction, the median survival was 11.2 to 43.4 months and the 5-years overall survival was 13 % to 23%. Important prognostic factors were found to be a low peritoneal carcarcinomatosis index (PCI) score and the completeness of cytoreduction. Conclusion: The current evidence suggests that CRS and HIPEC has a role to play in the treatment of peritoneal carcinomatosis from gastric cancer. Long term survival has been shown for a select group of patients. However, further studies are needed to validate these results.
Collapse
|
37
|
Heated IntraPEritoneal Chemotherapy (HIPEC) for Patients With Recurrent Ovarian Cancer: A Systematic Literature Review. Int J Gynecol Cancer 2016; 26:661-70. [DOI: 10.1097/igc.0000000000000664] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BackgroundDespite advances in surgical oncology, most patients with primary ovarian cancer develop a recurrence that is associated with a poor prognosis. The aim of this review was to establish the impact of Heated IntraPEritoneal Chemotherapy (HIPEC) in the overall survival of patients with recurrent ovarian cancer.MethodsA search of PubMed/MEDLINE databases was performed in February 2015 using the terms “recurrent ovarian cancer,” “cytoreductive surgery/cytoreduction,” and “heated/hyperthermic intraperitoneal chemotherapy.” Only English articles with available abstracts assessing the impact of HIPEC in patients with recurrent ovarian cancer were examined. The primary outcome measure was overall survival, whereas secondary outcomes included disease-free survival and HIPEC-related morbidity.ResultsSixteen studies with 1168 patients were analyzed. Most studies were Level IV, with 4 studies graded as Level III and 1 Level II. Cisplatin was the main chemotherapeutic agent used, but variations were observed in the actual technique, temperature of perfusate, and duration of treatment. In patients undergoing cytoreductive surgery and HIPEC, the overall survival ranged between 26.7 and 35 months, with disease-free survival varying between 8.5 and 48 months. Heated IntraPEritoneal Chemotherapy seems to confer survival benefits to patients with recurrent disease, with a randomized controlled study reporting that the overall survival is doubled when cytoreductive surgery is compared with cytoreductive surgery and chemotherapy (13. 4 vs 26.7 months). Heated IntraPEritoneal Chemotherapy–related morbidity ranged between 13.6% and 100%, but it was mainly minor and not significantly different from that experienced by patients who only underwent cytoreduction.ConclusionsCytoreductive surgery and HIPEC seem to be associated with promising results in patients with recurrent ovarian cancer. Large international prospective studies are required to further quantify the true efficacy of HIPEC and identify the optimal treatment protocol for a maximum survival benefit.
Collapse
|
38
|
Sugarbaker PH, Van der Speeten K. Surgical technology and pharmacology of hyperthermic perioperative chemotherapy. J Gastrointest Oncol 2016; 7:29-44. [PMID: 26941982 DOI: 10.3978/j.issn.2078-6891.2015.105] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although cytoreductive surgery (CRS) and hyperthermic perioperative chemotherapy (HIPEC) have not been shown to be effective by themselves, as a combined treatment they are now standard of care for peritoneal metastases from appendiceal cancer and from colorectal cancer as well as peritoneal mesothelioma. The timing of the HIPEC in relation to the CRS is crucial in that the HIPEC is to destroy minimal residual disease that remains following the CRS and prevent microscopic tumor emboli within the abdomen and pelvis from implanting within the resection site, within fibrinous clot, or within blood clot. Proper selection of chemotherapy agents is crucial to the long-term benefit of CRS and HIPEC. One must consider the response expected with the cancer chemotherapy agent, its area under the curve (AUC) ratio indicating the amount of dose intensity within the peritoneal space, and the drug retention within the peritoneal space for a prolonged exposure. Hyperthermia will augment the cytotoxicity of the cancer chemotherapy agents and improve drug penetration. Irrigation techniques should not be overlooked as an important means of reducing the cancer cell burden within the abdomen and pelvis. Multiple technologies for HIPEC exist and these have advantages and disadvantages. The techniques vary from a totally open technique with a vapor barrier over the open abdominal space to a totally closed technique whereby the HIPEC is administered at the completion of the surgical procedure. The open techniques depend on a table-mounted retractor for suspension of the skin edges allowing a reservoir to occur within the abdomen and pelvis. There are nearly a dozen commercially available hyperthermia pumps, all of which seem to perform adequately for HIPEC although there is a variable degree of convenience and documentation of the HIPEC procedure. As the management of peritoneal metastases has progressed over three decades, early cases are now seen in which a laparoscopic CRS and HIPEC may be appropriate. Also, prophylactic use of laparoscopic HIPEC with perforated appendiceal malignancies and T4 colon cancers may be appropriate.
Collapse
Affiliation(s)
- Paul H Sugarbaker
- 1 Center for Gastrointestinal Malignancies, MedStar Washington Hospital Center, Washington, DC, USA ; 2 Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Kurt Van der Speeten
- 1 Center for Gastrointestinal Malignancies, MedStar Washington Hospital Center, Washington, DC, USA ; 2 Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium
| |
Collapse
|
39
|
Hyperthermic Intraperitoneal Chemotherapy (HIPEC) Methodology, Drugs and Bidirectional Chemotherapy. Indian J Surg Oncol 2016; 7:152-9. [PMID: 27065705 DOI: 10.1007/s13193-016-0498-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 01/28/2016] [Indexed: 01/15/2023] Open
Abstract
Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) combined have been recognized as standard of care for treatment of a subset of patients with peritoneal carcinomatosis (PC). The aim of CRS is to eliminate all macroscopic disease through a series of visceral resections followed by targeting any residual microscopic disease with intraperitoneal chemotherapy, exposing the peritoneal surfaces to a high concentration of chemotherapy with a lower systemic toxicity. Different regimes of intraperitoneal chemotherapy include HIPEC, early postoperative intraperitoneal chemotherapy (EPIC) and bidirectional chemotherapy. The efficacy and modality of treatment with intraperitoneal chemotherapy is dependent on multiple factors including the chosen cytotoxic agent and its pharmacokinetics and pharmacodynamics. There is no standardized methodology for intraperitoneal chemotherapy administration. This review will discuss the pharmacological principles of the various intraperitoneal chemotherapy techniques.
Collapse
|
40
|
The Role of Hyperthermic Intraperitoneal Chemotherapy in Gastric Cancer. Indian J Surg Oncol 2016; 7:198-207. [PMID: 27065710 DOI: 10.1007/s13193-016-0502-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/28/2016] [Indexed: 12/26/2022] Open
Abstract
Peritoneal metastasis, either synchronous or metachronous, is commonly seen in gastric cancer. It is associated with a poor prognosis, with a median survival of less than one year. The outcomes are not significantly improved by the use of systemic chemotherapy. We review the relevant literature on the role of HIPEC in gastric cancer. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) has been used in three situations in gastric cancer. Besides its role as a definitive treatment in patients with established peritoneal metastasis (PM), it has been used as a prophylaxis against peritoneal recurrence after curative surgery and also as a palliative treatment in advanced peritoneal metastasis with intractable ascites. While prophylactic HIPEC has been shown to reduce peritoneal recurrence and improve survival in many randomised trials, palliative HIPEC can reduce the need for frequent paracentesis. Although CRS with HIPEC has shown promise in increasing the survival of selected patients with established PM from gastric cancer, larger studies are needed before this can be accepted as a standard of care.
Collapse
|
41
|
Chen Y, Li H, Jiang X, Chen D, Ni J, Sun H, Luo J, Yao H, Xu L. Regional thermochemotherapy versus hepatic arterial infusion chemotherapy for palliative treatment of advanced hilar cholangiocarcinoma: a retrospective controlled study. Eur Radiol 2016; 26:3500-9. [PMID: 26822373 DOI: 10.1007/s00330-016-4208-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 10/12/2015] [Accepted: 01/08/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To retrospectively assess the efficacy of regional thermochemotherapy (TCT) compared with hepatic arterial infusion chemotherapy (HAIC)-alone for palliative treatment of advanced hilar cholangiocarcinoma (HC) and to determine the prognostic factors associated with survival. METHODS Forty-three consecutive patients with advanced HC underwent regional TCT (TCT group) and HAIC (HAIC group). We analyzed baseline characteristics, overall survival (OS), progression-free survival (PFS), stent patency time (SPT), adverse events (AEs), and prognostic factors for OS between the two groups. RESULTS OS of patients treated with regional TCT was significantly longer compared to that of patients treated with HAIC (median OS: 20.3 vs. 13.2 months, P = 0.004), and SPT and PFS were significantly increased in the TCT group compared with the HAIC group (median SPT: 26.5 vs. 10.5 months, P < 0.001; median PFS: 16.5 vs. 10.2 months, P = 0.001). TCT and metal stent insertion were two independent prognostic factors associated with survival. The treatment-related AEs were tolerable and similar in the two groups, except for hilar pain (34.6 %) and skin rashes (24.6 %) in the TCT group. CONCLUSIONS Our results show that regional TCT is safe and more effective than HAIC-alone and may be a promising option for palliative treatment of advanced HC. Metal stenting before TCT appears to improve patients' OS. KEY POINTS • Regional TCT is a novel combination for palliative treatment of advanced HC • Our data showed significantly promising outcomes in the TCT group • HC patients with metal stenting appeared to derive greater benefit from TCT.
Collapse
Affiliation(s)
- Yaoting Chen
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Huiqing Li
- Health Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Xiongying Jiang
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Dong Chen
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Jiayan Ni
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Hongliang Sun
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Jianghong Luo
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China.
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China.
| | - Linfeng Xu
- Department of Interventional Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang Road West, Yuexiu Region, Guangzhou, Guangdong, 510120, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang Road West, Guangzhou, Guangdong, 510120, People's Republic of China.
| |
Collapse
|
42
|
Baratti D, Kusamura S, Pietrantonio F, Guaglio M, Niger M, Deraco M. Progress in treatments for colorectal cancer peritoneal metastases during the years 2010-2015. A systematic review. Crit Rev Oncol Hematol 2016; 100:209-22. [PMID: 26867984 DOI: 10.1016/j.critrevonc.2016.01.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/25/2015] [Accepted: 01/16/2016] [Indexed: 02/08/2023] Open
Abstract
Peritoneal metastases (PM) from colorectal cancer (CRC) were traditionally associated with bad prognosis. Only recently, cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) has resulted in survival improvements. A systematic literature search between January 2010 and June 2015 was performed. Studies were selected and appraised according to predetermined criteria. Nineteen cohort studies, and thirteen comparative studies of CRS/HIPEC were included. The weighted median overall survival was 31.6 months (range 16-51). Major morbidity was 17.6-52.4% (weighted average 32.6%). Mortality was 0-8.1% (weighted average 2.9%). Additional relevant topics, such as CRC-PM prevalence, results by systemic therapies, preoperative work-up, and technical aspects were summarized through a narrative review. The recent literature suggests that CRS/HIPEC is gaining acceptance as standard of care for selected CRC-PM patients. Refinement of selection criteria, and rationalization of comprehensive systemic and local-regional management is ongoing. Prevention and early treatment of PM are new and promising options.
Collapse
Affiliation(s)
- Dario Baratti
- Peritoneal Malignancy Program, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian, 1 20133 Milano, Italy
| | - Shigeki Kusamura
- Peritoneal Malignancy Program, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian, 1 20133 Milano, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian, 1 20133 Milano, Italy
| | - Marcello Guaglio
- Peritoneal Malignancy Program, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian, 1 20133 Milano, Italy
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian, 1 20133 Milano, Italy
| | - Marcello Deraco
- Peritoneal Malignancy Program, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian, 1 20133 Milano, Italy.
| |
Collapse
|
43
|
Seshadri RA, Glehen O. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in gastric cancer. World J Gastroenterol 2016; 22:1114-30. [PMID: 26811651 PMCID: PMC4716024 DOI: 10.3748/wjg.v22.i3.1114] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer associated peritoneal carcinomatosis (GCPC) has a poor prognosis with a median survival of less than one year. Systemic chemotherapy including targeted agents has not been found to significantly increase the survival in GCPC. Since recurrent gastric cancer remains confined to the abdominal cavity in many patients, regional therapies like aggressive cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) have been investigated for GCPC. HIPEC has been used for three indications in GC- as an adjuvant therapy after a curative surgery, HIPEC has been shown to improve survival and reduce peritoneal recurrences in many randomised trials in Asian countries; as a definitive treatment in established PC, HIPEC along with CRS is the only therapeutic modality that has resulted in long-term survival in select groups of patients; as a palliative treatment in advanced PC with intractable ascites, HIPEC has been shown to control ascites and reduce the need for frequent paracentesis. While the results of randomised trials of adjuvant HIPEC from western centres are awaited, the role of HIPEC in the treatment of GCPC is still evolving and needs larger studies before it is accepted as a standard of care.
Collapse
|
44
|
Ihemelandu C, Bijelic L, Sugarbaker PH. Iterative Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Recurrent or Progressive Diffuse Malignant Peritoneal Mesothelioma: Clinicopathologic Characteristics and Survival Outcome. Ann Surg Oncol 2015; 22:1680-1685. [DOI: 10.1245/s10434-014-3977-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
45
|
Budker VG, Monahan SD, Subbotin VM. Loco-regional cancer drug therapy: present approaches and rapidly reversible hydrophobization (RRH) of therapeutic agents as the future direction. Drug Discov Today 2014; 19:1855-70. [PMID: 25173702 DOI: 10.1016/j.drudis.2014.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/10/2014] [Accepted: 08/21/2014] [Indexed: 01/01/2023]
Abstract
Insufficient drug uptake by solid tumors remains the major problem for systemic chemotherapy. Many studies have demonstrated anticancer drug effects to be dose-dependent, although dose-escalation studies have resulted in limited survival benefit with increased systemic toxicities. One solution to this has been the idea of loco-regional drug treatments, which offer dramatically higher drug concentrations in tumor tissues while minimizing systemic toxicity. Although loco-regional delivery has been most prominent in cancers of the liver, soft tissues and serosal peritoneal malignancies, survival benefits are very far from desirable. This review discusses the evolution of loco-regional treatments, the present approaches and offers rapidly reversible hydrophobization of drugs as the new future direction.
Collapse
|
46
|
Blackham AU, Russell GB, Stewart JH, Votanopoulos K, Levine EA, Shen P. Metastatic colorectal cancer: survival comparison of hepatic resection versus cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg Oncol 2014; 21:2667-74. [PMID: 24615177 DOI: 10.1245/s10434-014-3563-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Liver resection has long been considered the standard of care for resectable colorectal hepatic metastases (HM). Patients with colorectal peritoneal surface disease (PSD) are now also being treated with aggressive therapy in the form of cytoreductive surgery (CS) and hyperthermic intraperitoneal chemotherapy (HIPEC). METHODS A retrospective comparison of optimally-treated colorectal cancer patients with HM or PSD obtained from prospectively maintained databases (1991-2010). RESULTS Liver resection was performed on 179 patients with HM, while 93 PSD patients received a complete cytoreduction followed by HIPEC. Patients differed in terms of age, performance status, site of primary cancer, T stage, and the use of perioperative chemotherapy. Five-year overall survival for HM patients was 36 %, with a median survival of 46 months, compared with 26 % and 34 months in patients with PSD (p = 0.024). When stratified by resection status, R0 HM (n = 170) and R0 PSD (n = 48) patients had similar median survival (49 vs. 41 months; p = 0.39). Median survival following R1 resection was also similar among HM (n = 9) and PSD (n = 45) patients (28 vs. 23 months; p = 0.68). Multivariate analysis identified distinctly different independent prognostic factors between HM and PSD patients. Major morbidity was 21 and 23 % (p = 0.88), while mortality was 3.9 versus 5.4 % (p = 0.55) in the HM and PSD patients, respectively. CONCLUSION Colorectal HM and PSD are distinct biologic diseases with different presentations and unique prognostic factors. However, long-term survival following CS/HIPEC is comparable to liver resection when stratified by completeness of resection. Furthermore, perioperative morbidity and mortality are similar.
Collapse
Affiliation(s)
- Aaron U Blackham
- Department of General Surgery, Surgical Oncology Section, Wake Forest School of Medicine, Winston-Salem, NC, USA,
| | | | | | | | | | | |
Collapse
|
47
|
Speeten KVD, Stuart AO, Sugarbaker PH. Pharmacology of cancer chemotherapy drugs for hyperthermic intraperitoneal peroperative chemotherapy in epithelial ovarian cancer. World J Obstet Gynecol 2013; 2:143-152. [DOI: 10.5317/wjog.v2.i4.143] [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: 12/13/2012] [Accepted: 07/05/2013] [Indexed: 02/05/2023] Open
Abstract
The peritoneal parietal and visceral surfaces of the abdomen and pelvis are an important anatomic site for the dissemination of epithelial ovarian cancer (EOC). The transcoelomic spread of cancer cells gives rise to peritoneal carcinomatosis (PC) which, without special treatments, is a fatal manifestation of EOC. In order to control PC cytoreductive surgery to remove macroscopic disease is combined with perioperative intraperitoneal (IP) and perioperative intravenous chemotherapy to eradicate microscopic residual disease. Chemotherapy agents are selected to be administered by the IP or intravenous route based on their pharmacologic properties. A peritoneal-plasma barrier which retards the clearance of high molecular weight chemotherapy from the peritoneal cavity results in a large exposure of small cancer nodules on abdominal and pelvic surfaces. Tissue penetration is facilitated by moderate hyperthermia (41-42 °C) of the IP chemotherapy solution. Timing of the chemotherapy as a planned part of the surgical procedure to maximize exposure of all peritoneal surfaces is crucial to success.
Collapse
|
48
|
Roviello F, Caruso S, Neri A, Marrelli D. Treatment and prevention of peritoneal carcinomatosis from gastric cancer by cytoreductive surgery and hyperthermic intraperitoneal chemotherapy: overview and rationale. Eur J Surg Oncol 2013; 39:1309-16. [PMID: 24183797 DOI: 10.1016/j.ejso.2013.10.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/19/2013] [Accepted: 10/09/2013] [Indexed: 12/24/2022] Open
Abstract
Peritoneal carcinomatosis (PC) from gastric cancer is a condition with a very bleak prognosis. Most authors consider it to be a terminal disease and recommend palliative therapy only. Multimodal therapeutic approaches to PC have emerged in the last decades, combining cytoreductive surgery (CRS) and peritonectomy procedures with perioperative intraperitoneal chemotherapy (IPEC), including hyperthermic intraperitoneal chemotherapy (HIPEC) and/or early postoperative intraperitoneal chemotherapy (EPIC). We reviewed the pertinent literature concerning the HIPEC modality both for the treatment of established PC and the prevention of peritoneal recurrence after potentially curative gastric cancer (GC) surgery. Basically, the two procedures relate to different aspects of GC and they are not comparable, since the latter has been used as an adjuvant when PC is still not macroscopically evident and the former has been exclusively used in advanced gastric cancer stages with peritoneal dissemination. Data supporting beneficial effects once gastric PC is already manifest is scarce and limited to few centres with specific experience in this field. Conversely, with regards to the peritoneal perfusion for preventing PC in high risk gastric cancer patients, there are phase III trials and meta-analysis which support beneficial effects resulting from the HIPEC procedure. To offer a baseline guide, we summarized the actual status and general outcome obtained by this multimodal technique, in association or not with CRS as treatment of advanced GC.
Collapse
Affiliation(s)
- F Roviello
- Department of Human Pathology and Oncology, Section of General Surgery and Surgical Oncology, University of Siena, Viale Bracci-Policlinico "Le Scotte", 53100 Siena, Italy
| | | | | | | |
Collapse
|
49
|
Gonzalez Bayon L, Steiner M, Vasquez Jimenez W, Asencio J, Alvarez de Sierra P, Atahualpa Arenas F, Rodriguez del Campo J, Garcia Sabrido J. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for the treatment of advanced epithelial ovarian carcinoma: Upfront therapy, at first recurrence, or later? Eur J Surg Oncol 2013; 39:1109-15. [DOI: 10.1016/j.ejso.2013.06.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/13/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022] Open
|
50
|
MacArthur KM, Nicholl MB. Principles and Innovations in Peritoneal Surface Malignancy Treatment. World J Oncol 2013; 4:129-136. [PMID: 29147344 PMCID: PMC5649776 DOI: 10.4021/wjon660w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2013] [Indexed: 12/29/2022] Open
Abstract
Cytoreductive surgery with heated intraperitoneal chemotherapy (CRS/HIPEC) remains a controversial treatment for malignant disease of the peritoneal cavity. We review the scientific principles underscoring the rationale for CRS/HIPEC, recent innovations and ongoing controversies. Lack of level 1 data limits the understanding of the true benefit of CRS/HIPEC.
Collapse
Affiliation(s)
- Kelly M MacArthur
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri 65212, USA
| | - Michael B Nicholl
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri 65212, USA.,Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, Missouri 65212, USA
| |
Collapse
|