1
|
Lapidot M, Sattler M. The Role of Surgery in Pleural Mesothelioma. Cancers (Basel) 2024; 16:1719. [PMID: 38730667 PMCID: PMC11083222 DOI: 10.3390/cancers16091719] [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: 03/27/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Surgery plays a central role in the diagnosis, staging, and management of pleural mesothelioma. Achieving an accurate diagnosis through surgical intervention and identifying the specific histologic subtype is crucial for determining the appropriate course of treatment. The histologic subtype guides decisions regarding the use of chemotherapy, immunotherapy, or multimodality treatment. The goal of surgery as part of multimodality treatment is to accomplish macroscopic complete resection with the eradication of grossly visible and palpable disease. Over the past two decades, many medical centers worldwide have shifted from performing extra-pleural pneumonectomy (EPP) to pleurectomy decortication (PD). This transition is motivated by the lower rates of short-term mortality and morbidity associated with PD and similar or even better long-term survival outcomes, compared to EPP. This review aims to outline the role of surgery in diagnosing, staging, and treating patients with pleural mesothelioma.
Collapse
Affiliation(s)
- Moshe Lapidot
- Division of Thoracic Surgery, Lung Center and International Mesothelioma Program, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Thoracic Surgery, Galilee Medical Center, Nahariya 2210001, Israel
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| |
Collapse
|
2
|
Miller DL, Parks CS, Ange B, Bonta IR, Rich PT. Hyperthermic intrathoracic extracorporeal chemotherapy for secondary malignant pleural disease. J Surg Oncol 2023; 128:604-611. [PMID: 37409778 DOI: 10.1002/jso.27389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/29/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
OBJECTIVES Pleural metastasis has extremely poor prognosis. Resection of pleural implants with infusion of intrathoracic hyperthermic chemotherapy may offer a survival advantage in selected patients. We evaluated the safety and efficacy of hyperthermic intrathoracic extracorporeal chemotherapy (HITEC) in patients who underwent pleurectomy/decortication (P/D) for secondary malignant pleural disease (SPD). METHODS A total of 101 patients were evaluated over 72 months, with 35 patients electing to proceed with P/D and 60 minutes of HITEC with cisplatin at 42°C. Inclusion criteria were adults 18-79 years with unilateral pleural dissemination. Exclusion criteria were patients without control of primary site, extrathoracic metastatic disease, significant comorbidities, and a history of adverse reaction to cisplatin. RESULTS Median age was 56 years (36-73); 60% were women. SPD was thymoma in 13, breast cancer in 9, lung cancer in 6, colon cancer in 2, renal cell in 2, and esophageal, anal, and thymic cancers in one each. There was no operative mortality. Postoperative complications occurred in 18 patients (51%). No patient developed renal failure. Median follow-up was 24 months (4-60). The overall survival rate was 61%; 17 patients (49%) developed recurrent disease at a median of 12 months (6-36). There were no recurrences after 36 months Eleven patients (31%) died of metastatic disease at a median of 17 months (7-25). CONCLUSIONS Surgical cytoreduction of SPD followed by HITEC with cisplatin was well tolerated. No patient developed cisplatin-related toxicities. Long-term follow-up is warranted to determine survival advantage and refinement of inclusion criteria.
Collapse
|
3
|
Lapidot M, Mazzola E, Bueno R. Outcomes of Pleurectomy Decortication in Biphasic Mesothelioma. J Thorac Cardiovasc Surg 2022; 164:1340-1348.e3. [DOI: 10.1016/j.jtcvs.2022.01.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 12/11/2021] [Accepted: 01/12/2022] [Indexed: 10/31/2022]
|
4
|
Habti M, Hubert G, Marzouki M, Piché N. Multimodal Treatment Including Intracavitary Chemotherapy for Recurrent Pediatric Pleuropulmonary Blastoma. Cureus 2021; 13:e20584. [PMID: 35103161 PMCID: PMC8782630 DOI: 10.7759/cureus.20584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 11/05/2022] Open
Abstract
Pleuropulmonary blastoma (PPB) is a rare pediatric cancer, and although there have been improvements in its treatment approach, recurrences retain a very poor prognosis. Here, we report a case of a 30-month-old female who survived recurrent PPB after undergoing surgery, adjuvant chemotherapy, intrapleural cisplatin infusion, and targeted therapy through whole exome sequencing (WES). Intrapleural cisplatin infusion and target therapy appear to be safe and can be considered in a multimodal approach for the management of recurrent PPB.
Collapse
|
5
|
Lapidot M, Mazzola E, Freyaldenhoven S, De León LE, Jaklitsch MT, Bueno R. Postoperative empyema after pleurectomy decortication for malignant pleural mesothelioma. Ann Thorac Surg 2021; 114:1214-1219. [PMID: 34619137 DOI: 10.1016/j.athoracsur.2021.08.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/11/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Postoperative empyema following pleurectomy decortication (PDC) for malignant pleural mesothelioma (MPM) is a serious complication that necessitates prolonged hospitalization. The aim of this study was to determine the incidence, risk factors and prognosis in patients who develop postoperative empyema following PDC. METHODS The background, type of PDC, neo-adjuvant treatment, date of empyema, pleural fluid cultures, post empyema treatment and prognosis from a series of consecutive 355 patients treated over 9 years at a single high-volume center were investigated. Fisher's exact test, Kaplan Meier estimators and log rank test were used to identify significant risk factors for postoperative empyema and compare the overall survival. RESULTS 355 patients underwent PDC for MPM in a 9-year period. There were 263 males and median age at surgery was 69. Neoadjuvant therapy was given to 87 and 282 received intraoperative heated chemotherapy (IOHC). During the study 24 patients (6.8%) developed empyema. The length of stay (LOS) of patients who developed postoperative empyema was significantly longer. Median survival for patients who developed postoperative empyema was 11.7 months and 21.3 months for patients without empyema (HR-1.78, p=0.009). Postoperative empyema was associated with male sex, prolonged air leak and use of prosthetic mesh. CONCLUSIONS Postoperative empyema following PDC is associated with prolonged length of stay and higher mortality. The rates of this serious postoperative complication might decrease by developing better strategies to avoid prolonged air leak after PDC.
Collapse
Affiliation(s)
- Moshe Lapidot
- Brigham & Women's Hospital, The International Mesothelioma Program, Harvard Medical School Boston, MA.
| | - Emanuele Mazzola
- Dana Farber Cancer Institute, T.H Chan School of Public Health Boston, Harvard Medical School Boston, MA
| | - Samuel Freyaldenhoven
- Brigham & Women's Hospital, The International Mesothelioma Program, Harvard Medical School Boston, MA
| | - Luis E De León
- Brigham & Women's Hospital, The International Mesothelioma Program, Harvard Medical School Boston, MA
| | - Michael T Jaklitsch
- Brigham & Women's Hospital, The International Mesothelioma Program, Harvard Medical School Boston, MA
| | - Raphael Bueno
- Brigham & Women's Hospital, The International Mesothelioma Program, Harvard Medical School Boston, MA
| |
Collapse
|
6
|
Pleurectomy Decortication in the Treatment of Malignant Pleural Mesothelioma: Encouraging Results and Novel Prognostic Implications Based on Experience in 355 Consecutive Patients. Ann Surg 2020; 275:1212-1220. [PMID: 33278174 DOI: 10.1097/sla.0000000000004306] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE We report a series of 355 consecutive patients treated over 9 years in a single institution with intended PDC. BACKGROUND Surgery for MPM has shifted from extra-pleural pneumonectomy to PDC with the goal of MCR. METHODS Clinical and outcome data were reviewed. Kaplan-Meier estimators and log rank test were used to compare the overall survival, and logistic regression models were used. RESULTS MCR was achieved in 304. There were 223 males, median age was 69 and histology was epithelioid in 184. The 30 and 90-day mortality were 3.0% and 4.6%.Most complications were low grade. Prolonged air leak in 141, deep venous thrombosis in 64, Atrial fibrillation in 42, chylothorax in 24, Empyema in 23, pneumonia in 21, Hemothorax in 12 and pulmonary embolus in 8.Median/5-year survival were 20.7 months/17.9% in the intent-to-treat cohort and 23.2 months/21.2% in the MCR group. The survivals were best for patients with T1stage and epithelioid histology (69.8 months/54.1%). In a multivariable analysis, factors that were found to be associated with longer patient overall survival included epithelioid histology, T stage, quantitative clinical stage/tumor volume staging, adjuvant chemotherapy, intraoperative heated chemo, female sex, and length of stay shorter than 14 days. CONCLUSIONS PDC is feasible with low mortality and is associated with manageable complication rates. 5-year survival of patients undergoing PDC with MCR in multi-modality setting is approaching 25% depending on quantitative and clinical stage, sex and histological subtype and is better than PDC without- MCR.
Collapse
|
7
|
Opitz I, Lauk O, Meerang M, Jetter A, Aeschlimann B, Seifert B, Günther D, Stahel RA, Weder W. Intracavitary cisplatin-fibrin chemotherapy after surgery for malignant pleural mesothelioma: A phase I trial. J Thorac Cardiovasc Surg 2019; 159:330-340.e4. [PMID: 31590949 DOI: 10.1016/j.jtcvs.2019.07.073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES Intracavitary chemotherapy is a promising concept to improve local tumor control for malignant pleural mesothelioma with reported high morbidity rates. We have demonstrated that administration of cisplatin loaded to fibrin increased local drug concentration and reduced systemic toxicity in preclinical models. We present a phase I trial of intracavitary cisplatin-fibrin after surgical tumor resection. METHODS A total of 12 patients (75% International Mesothelioma Interest Group stage III-IV) were treated with 4 dose levels of intracavitary cisplatin-fibrin (11-44 mg/m2 body surface area) in a dose-escalating design. Cisplatin-fibrin was sprayed on the resected surfaces after pleurectomy/decortication. Blood and tissue samples were taken to assess toxicity and pharmacokinetics. Patients were regularly followed up. RESULTS No dose-limiting toxicity was observed. Major morbidity occurred in 4 patients (33%). The 30-day and 90-day mortality were both 0%. Of 80 adverse events, 9 were classified serious, but none of these were related to study treatment. Local cisplatin concentration in the chest wall tissue was high at all dose levels (median, 46.3 μg/g [12-133 μg/g]). In serum, median cisplatin area under the concentration time curve values were always below renal toxicity levels. The median overall survival with 95% confidence interval was 21 months (10-31 months). In 1 patient with epithelioid malignant pleural mesothelioma (International Mesothelioma Interest Group stage I), there was no sign of relapse 48 months after treatment (44 mg/m2 body surface area). CONCLUSIONS The administration of intracavitary cisplatin-fibrin is safe with favorable pharmacokinetics. Although most patients had advanced disease, long-term outcomes are comparable to other multimodal concepts. A confirmation phase II trial is ongoing.
Collapse
Affiliation(s)
- Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland.
| | - Olivia Lauk
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Mayura Meerang
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Alexander Jetter
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Beat Aeschlimann
- Department of Chemistry and Applied Biosciences and Laboratory of Inorganic Chemistry, ETH Zurich, Zurich, Switzerland
| | - Burkhardt Seifert
- Department of Biostatistics, Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Detlef Günther
- Department of Chemistry and Applied Biosciences and Laboratory of Inorganic Chemistry, ETH Zurich, Zurich, Switzerland
| | - Rolf A Stahel
- Laboratory of Molecular Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Mahvi DA, Liu R, Grinstaff MW, Colson YL, Raut CP. Local Cancer Recurrence: The Realities, Challenges, and Opportunities for New Therapies. CA Cancer J Clin 2018; 68:488-505. [PMID: 30328620 PMCID: PMC6239861 DOI: 10.3322/caac.21498] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Locoregional recurrence negatively impacts both long-term survival and quality of life for several malignancies. For appropriate-risk patients with an isolated, resectable, local recurrence, surgery represents the only potentially curative therapy. However, oncologic outcomes remain inferior for patients with locally recurrent disease even after macroscopically complete resection. Unfortunately, these operations are often extensive, with significant perioperative morbidity and mortality. This review highlights selected malignancies (mesothelioma, sarcoma, lung cancer, breast cancer, rectal cancer, and peritoneal surface malignancies) in which surgical resection is a key treatment modality and local recurrence plays a significant role in overall oncologic outcome with regard to survival and quality of life. For each type of cancer, the current, state-of-the-art treatment strategies and their outcomes are assessed. The need for additional therapeutic options is presented given the limitations of the current standard therapies. New and emerging treatment modalities, including polymer films and nanoparticles, are highlighted as potential future solutions for both prevention and treatment of locally recurrent cancers. Finally, the authors identify additional clinical and research opportunities and propose future research strategies based on the various patterns of local recurrence among the different cancers.
Collapse
Affiliation(s)
- David A Mahvi
- Postdoctoral Research Fellow, Division of Surgical Oncology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rong Liu
- Instructor in Surgery, Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mark W Grinstaff
- Professor of Translational Research, Biomedical Engineering, Chemistry, Materials Science and Engineering, and Medicine, Department of Chemistry, Boston University, Boston, MA
| | - Yolonda L Colson
- Michael A. Bell Family Distinguished Chair in Healthcare Innovation and Professor of Surgery, Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Chandrajit P Raut
- Associate Professor of Surgery, Division of Surgical Oncology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Masuelli L, Benvenuto M, Mattera R, Di Stefano E, Zago E, Taffera G, Tresoldi I, Giganti MG, Frajese GV, Berardi G, Modesti A, Bei R. In Vitro and In Vivo Anti-tumoral Effects of the Flavonoid Apigenin in Malignant Mesothelioma. Front Pharmacol 2017; 8:373. [PMID: 28674496 PMCID: PMC5474957 DOI: 10.3389/fphar.2017.00373] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/30/2017] [Indexed: 01/29/2023] Open
Abstract
Malignant mesothelioma (MM) is a tumor arising from mesothelium. MM patients’ survival is poor. The polyphenol 4′,5,7,-trihydroxyflavone Apigenin (API) is a “multifunctional drug”. Several studies have demonstrated API anti-tumoral effects. However, little is known on the in vitro and in vivo anti-tumoral effects of API in MM. Thus, we analyzed the in vitro effects of API on cell proliferation, cell cycle regulation, pro-survival signaling pathways, apoptosis, and autophagy of human and mouse MM cells. We evaluated the in vivo anti-tumor activities of API in mice transplanted with MM #40a cells forming ascites. API inhibited in vitro MM cells survival, increased reactive oxygen species intracellular production and induced DNA damage. API activated apoptosis but not autophagy. API-induced apoptosis was sustained by the increase of Bax/Bcl-2 ratio, increase of p53 expression, activation of both caspase 9 and caspase 8, cleavage of PARP-1, and increase of the percentage of cells in subG1 phase. API treatment affected the phosphorylation of ERK1/2, JNK and p38 MAPKs in a cell-type specific manner, inhibited AKT phosphorylation, decreased c-Jun expression and phosphorylation, and inhibited NF-κB nuclear translocation. Intraperitoneal administration of API increased the median survival of C57BL/6 mice intraperitoneally transplanted with #40a cells and reduced the risk of tumor growth. Our findings may have important implications for the design of MM treatment using API.
Collapse
Affiliation(s)
- Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza",Rome, Italy
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy
| | - Rosanna Mattera
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy
| | - Enrica Di Stefano
- Department of Experimental Medicine, University of Rome "Sapienza",Rome, Italy
| | - Erika Zago
- Department of Experimental Medicine, University of Rome "Sapienza",Rome, Italy
| | - Gloria Taffera
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy
| | - Ilaria Tresoldi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy
| | - Giovanni Vanni Frajese
- Department of Sports Science, Human and Health, University of Rome "Foro Italico",Rome, Italy
| | - Ginevra Berardi
- Department of Chemistry, University of Rome "Sapienza",Rome, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy.,Center for Regenerative Medicine, University of Rome "Tor Vergata",Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata",Rome, Italy.,Center for Regenerative Medicine, University of Rome "Tor Vergata",Rome, Italy
| |
Collapse
|
10
|
Chan WH, Sugarbaker DJ, Burt BM. Intraoperative adjuncts for malignant pleural mesothelioma. Transl Lung Cancer Res 2017; 6:285-294. [PMID: 28713674 DOI: 10.21037/tlcr.2017.05.04] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rapidly fatal disease. Multimodality surgically based therapies may extend survival in select patients, however, local relapse after resection is common. Novel intraoperative adjunctive therapies including heated intraoperative chemotherapy (HIOC), heated intraoperative povidone-iodine (PVP-I), and photodynamic therapy (PDT) target micrometastatic disease and aim to improve local control. This review details the most recent studies and trials of HIOC, heated intraoperative PVP-I, and PDT, this aims to provide an update on some of the most promising intraoperative adjuncts for patients with MPM.
Collapse
Affiliation(s)
- Warren Ho Chan
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - David J Sugarbaker
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Bryan M Burt
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
11
|
Maury JM, Girard N, Tabutin M, Grima R, Chalabreysse L, Pavlakovic I, Sayag-Beaujard A, Leroux C, Souquet PJ, Glehen O, Tronc F. Intra-Thoracic Chemo-Hyperthermia for pleural recurrence of thymoma. Lung Cancer 2017. [PMID: 28625619 DOI: 10.1016/j.lungcan.2017.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Pleural recurrences are a hallmark of thymomas, and represent a challenge for multidisciplinary management. The purpose of this study was to assess the feasibility and the results in terms of morbidity, mortality and survival rates, of Intra-Thoracic Chemo-Hyperthermia (ITCH) for the treatment of pleural recurrences of thymomas. METHODS Retrospective analysis of 19 consecutives patients between 1997 and 2015 treated by surgical cytoreduction (pleurectomy) followed by ITCH with 25mg/m2 of mitomycin, and 50mg/m2 of Cisplatin. RESULTS There were 8 men and 11 women with a median age of 44 years. ITCH was combined with pleurectomy alone in 4 (22%) patients, pleurectomy and wedge resections in 14 (74%) patients; 1 (5%) patient had a pleuropneumonectomy. There were no perioperative deaths, and 5 patients (26%) presented with postoperative complication, including 3 (16%) cases related to chemotherapy (one case of reversible grade 2 bone marrow aplasia, and 2 cases of reversible, acute kidney failure). The median length of stay in intensive care unit and hospital were 1day and 10days, respectively. After a median follow-up period of 39 months (range 10-127 months), median disease-free survival was 42 months. Five patients (26%) died during follow-up. CONCLUSIONS Our data indicate that ITCH is a feasible option for selected patients with pleural recurrence of thymomas. ITCH clearly provides long local control, without major safety issues, and prolonged survival may be achieved in selected patients. This therapeutic option should be discussed at a multidisciplinary tumor board.
Collapse
Affiliation(s)
- Jean Michel Maury
- Department of Thoracic Surgery, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France; Université de Lyon, Université Lyon 1, INRA, UMR754, UMS 3444, SFR BioSciences, F-69007 Lyon, France
| | - Nicolas Girard
- Université de Lyon, Université Lyon 1, INRA, UMR754, UMS 3444, SFR BioSciences, F-69007 Lyon, France; Department of Respiratory Medicine, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France.
| | - Mayeul Tabutin
- Department of Thoracic Surgery, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France; Department of Surgery, Centre Léon-Berard, Cancer Research Center of Lyon 28, F-69008 Lyon, France
| | - Renaud Grima
- Department of Thoracic Surgery, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France
| | - Lara Chalabreysse
- Université de Lyon, Université Lyon 1, INRA, UMR754, UMS 3444, SFR BioSciences, F-69007 Lyon, France; Department of Pathology, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France
| | - Isabelle Pavlakovic
- Department of Anesthesiology and Reanimation, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France
| | - Annie Sayag-Beaujard
- Department of Anesthesiology and Reanimation, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France
| | - Caroline Leroux
- Université de Lyon, Université Lyon 1, INRA, UMR754, UMS 3444, SFR BioSciences, F-69007 Lyon, France
| | - Pierre-Jean Souquet
- Department of Respiratory Medicine, Lyon Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Benite, France
| | - Olivier Glehen
- Departement of General Surgery, Lyon Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Benite, France
| | - François Tronc
- Department of Thoracic Surgery, Louis Pradel Hospital, Hospices Civils de Lyon, F-69677 Lyon, France; Université de Lyon, Université Lyon 1, INRA, UMR754, UMS 3444, SFR BioSciences, F-69007 Lyon, France
| |
Collapse
|
12
|
Rosengart TK, Mason MC, LeMaire SA, Brandt ML, Coselli JS, Curley SA, Mattox KL, Mills JL, Sugarbaker DJ, Berger DA. The seven attributes of the academic surgeon: Critical aspects of the archetype and contributions to the surgical community. Am J Surg 2017; 214:165-179. [PMID: 28284432 DOI: 10.1016/j.amjsurg.2017.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 02/03/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND "Academic surgeon" describes a member of a medical school department of surgery, but this term does not fully define the important role of such physician-scientists in advancing surgical science through translational research and innovation. METHODS The curriculum vitae and self-descriptive vignettes of the records of achievement of seven surgeons possessing documented records of academic leadership, innovation, and dissemination of knowledge were reviewed. RESULTS Out analysis yielded seven attributes of the archetypal academic surgeon: 1) identifies complex clinical problems ignored or thought unsolvable by others, 2) becomes an expert, 3) innovates to advance treatment, 4) observes outcomes to further improve and innovate, 5) disseminates knowledge and expertise, 6) asks important questions to further improve care, and 7) trains the next generation of surgeons and scientists. CONCLUSION Although alternative pathways to innovation and academic contribution also exist, the academic surgeon typically devotes years of careful observation, analysis, and iterative investigation to identify and solve challenging or unexplored clinical problems, ideally leverages resources available in academic medical centers to support these endeavors.
Collapse
Affiliation(s)
- Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Meredith C Mason
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Scott A LeMaire
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Mary L Brandt
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Joseph S Coselli
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Steven A Curley
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Kenneth L Mattox
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Joseph L Mills
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - David J Sugarbaker
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - David A Berger
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
13
|
Gomez D, Tsao AS. Local and systemic therapies for malignant pleural mesothelioma. Curr Treat Options Oncol 2015; 15:683-99. [PMID: 25266654 DOI: 10.1007/s11864-014-0314-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OPINION STATEMENT Malignant pleural mesothelioma (MPM) is a challenging disease to treat with median overall survival times ranging between 9-17 months for all stages of disease. Recent clinical trials have improved our understanding of the biology of MPM. However, survival results are still not ideal. For early-stage MPM, patients should be evaluated for trimodality therapy in an experienced cancer center. If treating off-protocol, MPM patients should receive a surgical staging evaluation. The decision to proceed with surgical resection also should be considered after an extensive and thorough pulmonary and cardiac evaluation. If deemed a good surgical candidate, patients should receive surgical resection (pleurectomy/decortication or extrapleural pneumonectomy), adjuvant radiation therapy (hemithoracic external beam or intensity modulated radiation therapy), and either neoadjuvant or adjuvant chemotherapy (cisplatin-pemetrexed for 4 cycles). The optimal precise sequence of the trimodality is unclear and should be decided upon by a multidisciplinary consensus for each individual patient. In general, clinical trial participation should be encouraged. Several trials are currently underway to examine intraoperative therapies, vaccines, immunotherapy additions, and novel radiation therapy techniques. Advances in the field of MPM are reliant on participation in clinical trials and identifying biomarkers that are predictive for response to systemic therapies and prognostic for survival benefit.
Collapse
Affiliation(s)
- Daniel Gomez
- Department of Thoracic Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | | |
Collapse
|
14
|
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: 1.8] [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
|
15
|
Weder W, Opitz I. Multimodality therapy for malignant pleural mesothelioma. Ann Cardiothorac Surg 2013; 1:502-7. [PMID: 23977544 DOI: 10.3978/j.issn.2225-319x.2012.11.12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/22/2012] [Indexed: 12/20/2022]
Affiliation(s)
- Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | | |
Collapse
|
16
|
Abstract
Pleural malignancies are ideal for novel therapeutic approaches because they are invariably fatal. Intrapleural (IP) chemotherapy has only marginal benefit in pleural malignancies, but may prove efficacious with hyperthermic chemotherapy administered in combination with maximal tumor debulking. IP immunotherapies may be most effective in those patients with early-stage pleural malignancy, and may prove superior to standard pleurodesis methods in control of effusion and prolongation of survival. Immunogene therapy may be unable to successfully treat bulky tumors on its own, but success may be achieved with combination approaches that combine debulking surgery and chemotherapy with IP genetic immunotherapy.
Collapse
Affiliation(s)
- Andrew R Haas
- Section of Interventional Pulmonology and Thoracic Oncology, Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | | |
Collapse
|
17
|
Haas AR, Sterman DH. Malignant pleural mesothelioma: update on treatment options with a focus on novel therapies. Clin Chest Med 2013; 34:99-111. [PMID: 23411061 DOI: 10.1016/j.ccm.2012.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is evidence that improved treatments of malignant pleural mesothelioma are increasing the quality and quantity of life for patients with mesothelioma. Multimodality treatment programs that combine maximal surgical cytoreduction with novel forms of radiation therapy and more effective chemotherapy combinations may offer significant increases in survival for certain subgroups of patients with mesothelioma. Lung-sparing surgery may allow improvements in pulmonary function after surgery-based multimodality therapy, and potential longer overall survival than that seen with extrapleural pneumonectomy. Experimental treatments provide hope for all patients with mesothelioma, and in the future may be combined with standard therapy in multimodality protocols.
Collapse
Affiliation(s)
- Andrew R Haas
- Section of Interventional Pulmonology and Thoracic Oncology, Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Medical Center, 833 West Gates Building, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| | | |
Collapse
|
18
|
Facy O, Pages PB, Ortega-Deballon P, Magnin G, Ladoire S, Royer B, Chauffert B, Bernard A. High-pressure intrapleural chemotherapy: feasibility in the pig model. World J Surg Oncol 2012; 10:29. [PMID: 22309737 PMCID: PMC3395826 DOI: 10.1186/1477-7819-10-29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 02/06/2012] [Indexed: 12/12/2022] Open
Abstract
Background The usual treatments for pleural malignancies are mostly palliative. In contrast, peritoneal malignancies are often treated with a curative intent by cytoreductive surgery and intraperitoneal chemotherapy. As pressure has been shown to increase antitumor efficacy, we applied the concept of high-pressure intracavitary chemotherapy to the pleural space in a swine model. Methods Cisplatin and gemcitabine were selected because of their antineoplasic efficacy in vitro in a wide spectrum of cancer cell lines. The pleural cavity of 21 pigs was filled with saline solution; haemodynamic and respiratory parameters were monitored. The pressure was increased to 15-25 cm H2O. This treatment was associated with pneumonectomy in 6 pigs. Five pigs were treated with chemotherapy under pressure. Results The combination of gemcitabine (100 mg/l) and cisplatin (30 mg/l) was highly cytotoxic in vitro. The maximum tolerated pressure was 20 cm H20, due to haemodynamic failure. Pneumonectomy was not tolerated, either before or after pleural infusion. Five pigs survived intrapleural chemotherapy associating gemcitabine and cisplatin with 20 cm H2O pressure for 60 min. Conclusions High-pressure intrapleural chemotherapy is feasible in pigs. Further experiments will establish the pharmacokinetics and determine whether the benefit already shown in the peritoneum is also obtained in the pleura.
Collapse
|
19
|
Inhibition of cysteine peptidase activity in ascitic fluid in pancreatic cancer patients. Folia Histochem Cytobiol 2011; 48:513-7. [PMID: 21478091 DOI: 10.2478/v10042-010-0057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The work's objective is to answer the question whether there is any possibility of activity inhibition of cysteine peptidases inhibitors playing an important role in key processes accompanying cancer formation, including pancreas. There is a justified speculation that specific inhibitors of these enzymes may inhibit development of cancer processes by inhibiting their activity. In vitro studies confirmed that these enzymes in ascitic fluid were inhibited with egg whites inhibitors even to 90% of their original activity.
Collapse
|
20
|
Optimized intrapleural cisplatin chemotherapy with a fibrin carrier after extrapleural pneumonectomy: a preclinical study. J Thorac Cardiovasc Surg 2011; 141:65-71. [PMID: 21168013 DOI: 10.1016/j.jtcvs.2010.09.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 09/09/2010] [Accepted: 09/22/2010] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Our objective was to evaluate whether platinum concentrations in chest wall tissue and in serum are optimized by intracavitary application of cisplatin loaded to a fibrin carrier compared with cisplatin solution in a randomized setting of a pig model. METHODS After left-sided pneumonectomy including parietal pleurectomy, pigs were randomly assigned to receive either 90 mg/m(2) cisplatin intracavitary solution (n = 6) or to receive 5 mg cisplatin-fibrin (n = 5) applied on a predefined area of the chest wall. Platinum concentration in serum as well as in chest wall tissue was determined at several early time points until day 5 after treatment. Platinum levels were measured by inductively coupled plasma sector field mass spectrometric detection with a matrix-matched calibration procedure. RESULTS The dose- and surface-corrected (geometric) mean concentration of cisplatin in chest wall tissue 2 hours but also at day 5 after the application was doubled in animals treated with cisplatin-fibrin compared with the animals treated with cisplatin-solution. In serum, the dose- and surface-corrected exposure toward cisplatin (area under the curve(0-5d)) was significantly lower with cisplatin-fibrin than with cisplatin-solution (P < .0005). This is also reflected by significantly reduced serum creatinine and urea values in the cisplatin-fibrin group (P < .0001). Animals treated with cisplatin-fibrin additionally had a significantly better postoperative course as assessed by a well-being score (P < .001). CONCLUSIONS After cisplatin-fibrin treatment, cisplatin tissue concentration was increased whereas systemic cisplatin concentrations were significantly reduced in comparison with cisplatin-solution treatment. This finding offers a clear advantage inasmuch as rate and severity of systemic adverse events can be reduced while local cytotoxic concentrations are at least maintained.
Collapse
|
21
|
McAleer MF, Mehran RJ, Tsao A. Mesothelioma. Lung Cancer 2010. [DOI: 10.1007/978-1-60761-524-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
22
|
Su S. Mesothelioma: path to multimodality treatment. Semin Thorac Cardiovasc Surg 2009; 21:125-31. [PMID: 19822284 DOI: 10.1053/j.semtcvs.2009.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2009] [Indexed: 11/11/2022]
Abstract
Multimodality treatment of malignant pleural mesothelioma (MPM) with surgery, radiation therapy, and adjuvant or neoadjuvant chemotherapy is the sole path to extended survival for select patients with favorable prognostic factors. No single-modality approach has produced equivalent results. Much effort has been expended since first recognition of this insidious pleural cancer to elucidate the underlying cause and optimal treatment strategy. Despite recent breakthroughs, the principal barrier to achieving a cure rests with the propensity for disease recurrence in the ipsilateral hemithorax. Despite these limitations, however, the results hold promise for improved survival with further refinement in patient selection and targeted therapy. Other approaches to multimodality treatment have capitalized on an array of innovative technologies in search of the silver bullet strategy that will ultimately undermine the biological behavior demonstrated by MPM. These range from the use of different means of radiation delivery, biological agents, virally mediated gene therapy, photodynamic therapy, and immunotherapy. Additionally, studies using gene ratios will yield more accurate means by which to diagnose, distinguish prognosticators, and more selectively assign patients to aggressive treatments. In light of the current worldwide epidemic, the lessons learned over the past several decades serve as a humbling reminder of the treatment barriers that remain.
Collapse
Affiliation(s)
- Stacey Su
- Department of Surgery, University of Pennsylvania School of Medicine, University of Pennsylvania Health System, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
23
|
Tsao AS, Mehran R, Roth JA. Neoadjuvant and intrapleural therapies for malignant pleural mesothelioma. Clin Lung Cancer 2009; 10:36-41. [PMID: 19289370 DOI: 10.3816/clc.2009.n.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Historically, malignant pleural mesothelioma patients with potentially resectable disease have been treated with surgery and radiation alone. With improvements in systemic and intrapleural treatment options, a movement toward multi-modality therapy has become more common. Systemic treatment options largely consist of neoadjuvant chemotherapy with platinum doublets and most recently novel targeted agents, such as dasatinib. Intrapleural strategies have included injecting chemotherapy, chemotherapy with hyper thermic per fusion, gene therapy, and immunotherapy. The following review discusses the latest results in neoadjuvant and intrapleural therapies in malignant pleural mesothelioma.
Collapse
Affiliation(s)
- Anne S Tsao
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA.
| | | | | |
Collapse
|
24
|
Zellos L, Richards WG, Capalbo L, Jaklitsch MT, Chirieac LR, Johnson BE, Bueno R, Sugarbaker DJ. A phase I study of extrapleural pneumonectomy and intracavitary intraoperative hyperthermic cisplatin with amifostine cytoprotection for malignant pleural mesothelioma. J Thorac Cardiovasc Surg 2009; 137:453-8. [PMID: 19185169 DOI: 10.1016/j.jtcvs.2008.07.055] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 06/06/2008] [Accepted: 07/17/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE This study was undertaken to determine maximum tolerated dose and toxicity of intraoperative intracavitary hyperthermic cisplatin perfusion with amifostine after extrapleural pneumonectomy for malignant pleural mesothelioma. METHODS Patients with mesothelioma were prospectively enrolled. Those with resectable disease received amifostine and 1-hour hyperthermic cisplatin perfusion of ipsilateral hemithorax and abdomen. Morbidity, recurrence, and survival were recorded. RESULTS Forty-two patients were enrolled; 29 underwent resection (operative mortality 7%, 2/29). Median age was 57 years. Eighteen were in pathologic stage I or II; 11 were in stage III. Median hospitalization was 15 days. Common complications were atrial fibrillation (66%, 19 patients), deep venous thrombosis (31%, 9 patients), and grade 3+ renal toxicity (31%, 9 patients). Feasibility was determined. Renal toxicity was unrelated to cisplatin dose, with no maximum tolerated dose determined. Overall median survival was 17 months (resected 20 months, unresected 10 months). Median survivals were 26 months for patients receiving higher cisplatin doses and 16 months for those receiving lower doses (P = .35). Survival was significantly longer with negative extrapleural nodes (31 vs 14 months, P = .0115) and early stage (all resected 35 months for stage I-II vs 14 months for stage III, P = .0022, epithelial 39 months for stage I-II vs 15 months for stage III, P = .0072). CONCLUSION Early stage and negative extrapleural lymph nodes were associated with prolonged survival. Single-dose amifostine did not protect adequately against cisplatin-induced renal toxicity. Additional cytoprotective strategies are needed to allow determination of cisplatin maximum tolerated dose.
Collapse
Affiliation(s)
- Lambros Zellos
- Division of Thoracic Surgery and Department of Pathology, Brigham and Women's Hospital, Boston, Mass, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Mujoomdar AA, Sugarbaker DJ. Hyperthermic Chemoperfusion for the Treatment of Malignant Pleural Mesothelioma. Semin Thorac Cardiovasc Surg 2008; 20:298-304. [DOI: 10.1053/j.semtcvs.2008.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2008] [Indexed: 11/11/2022]
|
26
|
Anesthetic management of patients undergoing extrapleural pneumonectomy for mesothelioma. Curr Opin Anaesthesiol 2008; 21:21-7. [PMID: 18195605 DOI: 10.1097/aco.0b013e3282f2a9c3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Extrapleural pneumonectomy is a radical and aggressive surgery that presents a great challenge to the thoracic anesthesiologist. This surgery is performed routinely by only a few centers in the world and this review represents our institution's experience in anesthetic care. RECENT FINDINGS Prominent among the developing multimodal treatment options is the combination of extrapleural pneumonectomy with intraoperative intracavitary hyperthermic chemotherapy. Outcome survival benefits have recently been demonstrated for the less completely cytoreductive pleurectomy procedure when combined with intraoperative intracavitary hyperthermic chemotherapy and trials are well under way for extrapleural pneumonectomy plus intraoperative intracavitary hyperthermic chemotherapy. Anesthetic management of extrapleural pneumonectomy is further impacted by these developments. SUMMARY Anesthetic management importantly contributes to containment of the perioperative complications of extrapleural pneumonectomy. An appreciation of the technical aspects and physiologic disruptions associated with extrapleural pneumonectomy is critical to effective management. While data on this relatively uncommon surgical procedure are scarce, some referral centers have accumulated extensive experience. This review summarizes relevant surgical aspects and anesthetic insights from the Brigham and Women's Hospital experience. Included are the anesthetic implications of intraoperative intracavitary hyperthermic chemotherapy in combination with extrapleural pneumonectomy - an emerging therapeutic option in the treatment of malignant pleural mesothelioma.
Collapse
|
27
|
Schipper PH, Nichols FC, Thomse KM, Deschamps C, Cassivi SD, Allen MS, Pairolero PC. Malignant Pleural Mesothelioma: Surgical Management in 285 Patients. Ann Thorac Surg 2008; 85:257-64; discussion 264. [PMID: 18154820 DOI: 10.1016/j.athoracsur.2007.06.066] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 06/20/2007] [Accepted: 06/22/2007] [Indexed: 11/20/2022]
Affiliation(s)
- Paul H Schipper
- Division of General Thoracic Surgery, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Malignant pleural mesothelioma (MPM) is a resistant form of lung cancer that is often related to prior asbestos exposure. While surgical resection and radiotherapy techniques have been refined in recent years, neither has been proven to significantly extend patient survival compared with untreated controls. Until the release of pemetrexed in 2004, even combination chemotherapy regimens often resulted in a response rate of <20%. A recent phase III trial documented a 41.3% response rate for cisplatin plus pemetrexed. In the future, new multimodality regimens featuring novel targeted therapies directed against molecular targets, such as the vascular endothelial growth factor, hold the greatest promise for improved outcomes in MPM. The standard radiographic assessment of response to MPM therapy remains a poor surrogate for clinically relevant endpoints such as median survival. Furthermore, it is not currently known whether aggressive multimodality treatment for MPM will improve survival or quality of life above and beyond symptomatic care. Ongoing clinical trials are comparing chemotherapy and surgery with supportive care in an effort to define the role of different therapies in MPM. MPM treatment is a costly public health issue; after efficacy is proven, additional studies are needed to measure the cost effectiveness of MPM treatment regimens.
Collapse
Affiliation(s)
- Ranjit K Goudar
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27701, USA.
| |
Collapse
|
29
|
Johnstone DW. Re: Modified intrapleural cisplatin treatment for lung cancer with positive pleural lavage cytology or malignant effusion, by Muraoka M, Oka T, Akamine S, et al. J Surg Oncol 2006; 93:255-6. [PMID: 16496362 DOI: 10.1002/jso.20471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
30
|
Abstract
Mesothelioma is an extraordinarily challenging disease to treat. While numerous clinical trials testing various chemotherapeutic agents have been conducted over the last several decades, only recently have larger studies proven the efficacy of newer chemotherapy regimens. This article reviews the data regarding specific classes of chemotherapeutic agents. The role of treatment in various disease settings and the difficulty in assessing the benefit of that therapy is discussed. Finally, an update is provided on novel therapeutics being testing in mesothelioma.
Collapse
Affiliation(s)
- Lee M Krug
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
31
|
Abstract
Malignant pleural mesothelioma is an aggressive thoracic tumor that is often refractory to chemotherapy and radiotherapy, making long-term survival a difficult goal. Although multimodality therapies, including cytoreductive surgery, chemotherapy, and radiotherapy, have been tested, their survival benefit is unclear. Recently, novel cytotoxic agents such as pemetrexed have shown promise in the treatment of malignant pleural mesothelioma, and in early trials new small-molecule agents targeted to the specific biochemical properties of this tumor have shown promise.
Collapse
Affiliation(s)
- Ranjit Kumar Goudar
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|