Find information on thousands of medical conditions and prescription drugs.

Mesothelioma

Mesothelioma is an uncommon form of cancer, usually associated with previous exposure to asbestos. In this disease, malignant (cancerous) cells develop in the mesothelium, a protective lining that covers most of the body's internal organs. Its most common site is the pleura (outer lining of the lungs and chest cavity), but it may also occur in the peritoneum (the lining of the abdominal cavity) or the pericardium (a sac that surrounds the heart). more...

Home
Diseases
A
B
C
D
E
F
G
H
I
J
K
L
M
Mac Ardle disease
Macroglobulinemia
Macular degeneration
Mad cow disease
Maghazaji syndrome
Mal de debarquement
Malaria
Malignant hyperthermia
Mallory-Weiss syndrome
Malouf syndrome
Mannosidosis
Marburg fever
Marfan syndrome
MASA syndrome
Mast cell disease
Mastigophobia
Mastocytosis
Mastoiditis
MAT deficiency
Maturity onset diabetes...
McArdle disease
McCune-Albright syndrome
Measles
Mediterranean fever
Megaloblastic anemia
MELAS
Meleda Disease
Melioidosis
Melkersson-Rosenthal...
Melophobia
Meniere's disease
Meningioma
Meningitis
Mental retardation
Mercury (element)
Mesothelioma
Metabolic acidosis
Metabolic disorder
Metachondromatosis
Methylmalonic acidemia
Microcephaly
Microphobia
Microphthalmia
Microscopic polyangiitis
Microsporidiosis
Microtia, meatal atresia...
Migraine
Miller-Dieker syndrome
Mitochondrial Diseases
Mitochondrial...
Mitral valve prolapse
Mobius syndrome
MODY syndrome
Moebius syndrome
Molluscum contagiosum
MOMO syndrome
Mondini Dysplasia
Mondor's disease
Monoclonal gammopathy of...
Morquio syndrome
Motor neuron disease
Motorphobia
Moyamoya disease
MPO deficiency
MR
Mucopolysaccharidosis
Mucopolysaccharidosis...
Mullerian agenesis
Multiple chemical...
Multiple endocrine...
Multiple hereditary...
Multiple myeloma
Multiple organ failure
Multiple sclerosis
Multiple system atrophy
Mumps
Muscular dystrophy
Myalgic encephalomyelitis
Myasthenia gravis
Mycetoma
Mycophobia
Mycosis fungoides
Myelitis
Myelodysplasia
Myelodysplastic syndromes
Myelofibrosis
Myeloperoxidase deficiency
Myoadenylate deaminase...
Myocarditis
Myoclonus
Myoglobinuria
Myopathy
Myopia
Myositis
Myositis ossificans
Myxedema
Myxozoa
N
O
P
Q
R
S
T
U
V
W
X
Y
Z
Medicines

Most people who develop mesothelioma have worked on jobs where they inhaled asbestos particles, or have been exposed to asbestos dust and fibre in other ways, such as by washing the clothes of a family member who worked with asbestos, or by home renovation using asbestos cement products.

Signs and symptoms

Symptoms of mesothelioma may not appear until 30 to 50 years after exposure to asbestos. Shortness of breath and pain in the chest due to an accumulation of fluid in the pleural space are often symptoms of pleural mesothelioma.

Symptoms of peritoneal mesothelioma include weight loss and cachexia, abdominal swelling and pain due to ascites (a buildup of fluid in the abdominal cavity). Other symptoms of peritoneal mesothelioma may include bowel obstruction, blood clotting abnormalities, anemia, and fever. If the cancer has spread beyond the mesothelium to other parts of the body, symptoms may include pain, trouble swallowing, or swelling of the neck or face.

These symptoms may be caused by mesothelioma or by other, less serious conditions.

Diagnosis

Diagnosing mesothelioma is often difficult, because the symptoms are similar to those of a number of other conditions. Diagnosis begins with a review of the patient's medical history. A history of occupational exposure to asbestos may increase clinical suspicion for mesothelioma. A physical examination is performed, followed by chest X-ray and often lung function tests. The X-ray may reveal pleural thickening commonly seen after asbestos exposure and increases suspicion of mesothelioma. A CT (or CAT) scan or an MRI is usually performed. If a large amount of fluid is present, abnormal cells may be detected by cytology if this fluid is aspirated with a syringe. For pleural fluid this is done by a pleural tap or chest drain, in ascites with an paracentesis or ascitic drain and in a pericardial effusion with pericardiocentesis. While absence of malignant cells on cytology does not completely exclude mesothelioma, it makes it much more unlikely, especially if an alternative diagnosis can be made (e.g. tuberculosis, heart failure).

If cytology is positive or a plaque is regarded as suspicious, a biopsy is needed to confirm a diagnosis of mesothelioma. A doctor removes a sample of tissue for examination under a microscope by a histopathologist. A biopsy may be done in different ways, depending on where the abnormal area is located. If the cancer is in the chest, the doctor may perform a thoracoscopy. In this procedure, the doctor makes a small cut through the chest wall and puts a thin, lighted tube called a thoracoscope into the chest between two ribs. Thoracoscopy allows the doctor to look inside the chest and obtain tissue samples.

Read more at Wikipedia.org


[List your site here Free!]


Surgical treatment of malignant pleural mesothelioma : a review
From CHEST, 2/1/03 by Serge van Ruth

Despite many years of clinical research, there is still no effective therapy for malignant pleural mesothelioma (MPM). Untreated, the prognosis is poor, with a median survival of < 1 year. Single-agent or combination chemotherapy as well as radiotherapy have not shown persistent improvements in response or survival. In general, MPM is a disease confined to the pleural cavity for a long time before metastasizing. Therefore, focus on local treatment seems rational. Surgical resection has been considered the mainstay of treatment by some. However, surgery alone results in high recurrence rates, and the survival benefit remains questionable. In recent years, the emphasis has been on surgery combined with adjuvant therapies. In this article, the present state of surgical management of MPM will be reviewed.

Key words: adjuvant therapy; mesothelioma; pleural cavity; review; surgery

Abbreviations: EPP = extrapleural pneumonectomy; IMIG = International Mesothelioma Interest Group; MPM = malignant pleural mesothelioma; PDT = photodynamic therapy

**********

Malignant pleural mesothelioma (MPM) is an aggressive tumor of the pleura. Presenting symptoms are dyspnea and chest pain in the majority of patients. Coughing, fatigue, and weight loss are less frequently observed. (1,2) In general, MPM is a disease confined to the pleural cavity for a long time before metastasizing. (3) The most common features are pleural thickening, nodularity, and pleural effusion. The growth pattern is characterized by involving the entire pleura and interlobular space. (2) Malignant seeding along tracts of cytology or biopsy needles, chest tubes, thoracoscopy trocars, and surgical incisions is a common complication of diagnostic and therapeutic procedures in patients with MPM. (4) In Western Europe, 5,000 patients die of mesothelioma each year. (5) In the last decades, the incidence has increased twofold in the Netherlands, and it is expected to reach its maximum in the year 2020. (6) The association with asbestosis is well known. In approximately 80% of MPM, an exposure to asbestos is reported. The latency period is between 20 years and 30 years. (7,8) Recently, a virus has become a suspected agent too. (9) Simian virus 40, a DNA tumor virus, has the potential to induce mesothelioma in hamsters and is reported to be identified in a number of patients with MPM. (9) However, there are still discussions ongoing about the potential of simian virus 40 to induce MPM in humans. The prognosis of patients with MPM is poor; untreated, the median survival is 9 months. (10,11)

Systemic chemotherapy results in partial responses of between 15% and 20%; complete responses are rare. (12-15) Radiotherapy as single treatment modality, administered with curative intent, is considered not feasible because of the large target volume and the dose-limiting toxicity of the adjacent organs and structures. Radiotherapy is considered useful for palliation and prevention of seeding after invasive diagnostic procedures. (4,16,17)

Surgical resection has been considered the mainstay of treatment by some. However, it is almost impossible to achieve a microscopically complete resection with surgery alone because of the anatomy of the pleura and the property of MPM to infiltrate the underlying and neighboring structures. (18) Surgery alone is associated with a high recurrence rate. Recently, most efforts have been put in the combination of cytoreductive surgery with some form of adjuvant therapy (19,20)

In this article, we review the surgical management of MPM. The different techniques and treatment outcome of surgery alone are described. Thereafter, emphasis is given to the adjuvant therapies.

MATERIALS AND METHODS

A systemic literature study was performed to identify all relevant articles until October 2001. A MEDLINE search was performed with key words focused on MPM. Studies with < 10 patients were not included unless they showed very interesting results. When there were several reports of the same institute including the same cohort of patients with the same treatment, we listed only the most recent report. A statistical analysis of all reviewed articles was not possible due to the lack of randomized studies, the small patient groups, and the diversity of patient groups and methods.

Staging

Staging is important in the treatment of MPM. (21) Different staging systems are used (Table 1). (22-24) To stage accurately, several staging methods are used. Thoracoscopy, CT, MRI, and laparoscopy can identify the T status. (25,26) CT compared to MRI has nearly equivalent diagnostic accuracy. MRI is superior in imaging diaphragmatic muscle involvement, endothoracic involvement, and revealing solitary foci of chest wall invasion. (27) To accurately determine the nodal status is more often a problem. CT has a low accuracy regarding lymph nodes. (27) Mediastinoscopy is useful; however, 25% of the patients with MPM have nodal involvement confined to areas such as peridiaphragmatic and internal mammary regions not accessible to the mediastinoscopy. (28) Positron emission tomography seems to be useful to determine the extent of tumor. (29) Unfortunately, correct staging is only possible during operation in a substantial number of patients. The accuracy of preoperative CT scans to determine the stage correctly varies, but is reported as low as 30%. (1,30) The intraoperative tumor load is associated with outcome of MPM, and large volumes are associated with nodal spread. (31)

Prognostic Factors

In studies of Rusch and Venkatraman (28) and Sugarbaker et al, (32) the stage, histology, and adjuvant therapy, but not type of resection, were significant prognostic factors. Stage is a clear prognostic factor. Rusch and Venkatraman (28) reported a median survival after surgery with adjuvant therapy of 29.9 months for stage I, 19 months for stage II, 10.4 months for stage III, and 8 months for stage IV (International Mesothelioma Interest Group [IMIG] staging). Another study showed that when the visceral pleura was intact, the median survival was 32.7 months. (33) The node status alone has also prognostic significance with survival advantage for lymph node-negative patients. (23) Sarcomatous MPM shows a worse survival than the epithelial type. (23,32) Rusch and Venkatraman (28) found that female patients show better survival than male patients; however, Sugarbaker et al (32) could not confirm this. The type of resection, ie, extrapleural pneumonectomy (EPP) or a pleurectomy/decortication, did not have impact on survival in the study of Rusch and Venkatraman. (28) However, both procedures were performed only if they led to complete resection of all gross tumor. In patients with bulky tumor or confluent pleural tumor, an EPP was necessary to achieve complete resection. (28)

RESULTS

Surgery Alone

Pleurectomy/Decortication: The technique of pleurectomy has been well described. (34) After a posterolateral thoracotomy, an extrapleural plane between the parietal pleura and the endothoracic fascia is entered. The dissection proceeds in a superior direction toward the apex over the posterolateral aspect of the chest wall. The dissection is continued to inferior and posterior. When the pleura and the lung are completely mobilized in the upper part of the thoracic cavity, the superior and posterior hilar structures of the lung are well exposed. After stripping or partial resection of the posterior pericardium, the dissection proceeds toward the posterior diaphragmatic sulcus. If there is only superficial involvement, dissection is performed through the diaphragmatic muscle, avoiding entering the abdomen; otherwise, a part of the diaphragm is removed. The en bloc specimen is mobilized back to the pericardium medially. When the dissection is completed to the hilar structures, the parietal pleural is opened and decortication of the visceral pleura is performed. The pericardium and diaphragm are eventually reconstructed.

The mortality of this procedure is limited (1 to 2%), when performed in specialized centers. (34,35) The most common complication is prolonged air leakage, occurring in 10% of cases. Other reported complications are pneumonia, empyema, and hemorrhage. (2) Pleurectomy and decortication are reported to be effective in controlling pleural effusion. The median survival reached by this procedure is reported in different studies between 9 months and 20 months (Table 2).

The technical problem is the difficulty of separating the visceral pleura from the lung parenchyma. This results frequently in incomplete resection. (3) After pleurectomy/decortication, Hilaris et al (44) reported that residual tumor was left behind in 78% of the patients, most frequently on the visceral pleura. The most common site of recurrence is the ipsilateral hemithorax. (34) In recent years, pleurectomy/ decortication studies all included adjuvant therapy.

EPP: EPP is a procedure consisting of en bloc resection of the lung, visceral and pleural pleura, pericardium, and ipsilateral diaphragm with reconstruction of the pericardium and diaphragm. (45) After a posterolateral thoracotomy through the sixth intercostal space, a dissection between the chest wall and parietal pleura is started. A blunt dissection with fingers appears to work best. After reaching the apex of the chest, the dissection will be proceeded to inferior (diaphragm). The diaphragm is opened while aiming to preserve the peritoneum. The whole diaphragm is removed. Next, the pericardium is resected. The specimen is then elevated, and the dissection continues to the hilar structures. After stapling the vessels and the bronchus, the specimen is removed. A pericardial fat pad can be placed over the bronchial stump. Reconstruction of the diaphragm and pericardium is the last stage of the procedure. In the patch to reconstruct the pericardium, fenestrations are made to prevent cardiac tamponade.

The mortality of this procedure has decreased in the last decades from 30 to < 5% when performed in specialized centers and in selected patients. (46,47) Causes of death are respiratory failure, myocardial failure, and pulmonary embolus. (2) The reported morbidity is considerable, mostly between 25% and 50%. (15,46) Twenty-four percent of the patients undergoing pneumonectomy showed cardiac supraventricular dysrhythmias with a peak incidence at 3 to 4 days after resection. (48) Patients are at risk of postoperative pneumonia, and the development of a bronchopleural fistula is reported in 10 to 20%, especially right-sided EPP. (2,23) Median survival after EPP is ranges from 9 to 19 months (Table 3).

EPP is performed for locally advanced disease, usually in patients with confluent visceral pleural tumor not separable from the lung and a partially or totally fused pleural space. Compared with pleurectomy/decortication, a lower recurrence rate has been reported (10% after EPP vs 52% after pleurectomy). (41,50) However, relapses in distant sites are more frequently seen than in the pleurectomy group, especially in adjacent cavities.51 Because of operative deaths, residual tumor, local recurrence, and metastatic disease, EPP has not gained wide acceptance as treatment on its own. (41) There does not seem to be a survival benefit for patients undergoing EPP in comparison to patients undergoing pleurectomy. (41)

Surgery alone is associated with a high recurrence rate, and therefore adjuvant therapy seems useful. (19,20) Studies performed with the combination of surgery and adjuvant treatment are listed in Table 4.

Surgery and Emphasis on External Radiotherapy

In Table 4, series are collected that report on combination therapy of surgery with complete hemithoracic irradiation. Sugarbaker et al (67) advocated that adjuvant radiotherapy should be 40 to 45 Gy to the entire hemithorax, with a 5- to 5.5-Gy boost to areas at high risk for recurrence. Doses limiting thoracic structures are spinal cord (45 Gy), heart (45 Gy), and lung (20 Gy). (68) Hemithoracic radiotherapy equals a total loss of lung function. (69) A shift of the abdominal viscera into the inferior hemithorax after a pneumonectomy limits the safe dose to 30 Gy in the inferior area. (67)

The technique of EPP combined with hemithoracic radiation and systemic chemotherapy was described by Grondin and Sugarbaker. (70) The largest series was described by Sugarbaker et al (46) with 183 patients. The mortality rate was 3.8%. The morbidity rate was 50%, including cardiac arrest, respiratory failure, ARDS, sepsis, contralateral pneumothorax, arrhythmias, pulmonary embolism, empyema, and GI hemorrhage. (46) The median survival in this patient group was 19 months, with a 2-year survival of 38%. In selected patients with the epithelial cell type and without mediastinal nodal metastases at resection, Sugarbaker et al (67) reported a 5-year survival of 45%. Despite aggressive local treatment including pericardium and diaphragm resection, the site of failure was in most instances the ipsilateral hemithorax (35%) followed by the abdomen (26%), the contralateral hemithorax (17%), and other distant sites (8%). (50)

The application of brachytherapy after pleurectomy was studied in 41 patients by Hilaris et al. (44) Measurable gross residual tumor was treated with permanent iodine 125 implantation and residual diffuse disease by temporary iridium 92 implantation or postoperative instillation of phosphorus 32. After this treatment, external radiotherapy on the hemithorax was administered (45 Gy). There was no mortality. Complications occurred in six patients (15%), including one case of radiation pneumonitis and one case of pericarditis. The median survival was 21 months, with a 2-year survival of 40%. At time of the report, 71% of the patients had relapsed. Local recurrence occurred in one third of the relapsed patients, and distant metastasis with or without local recurrence occurred in the other two thirds. (44) An update, including the same patient cohort with larger follow-up, by Mychalczak et al (71) could not confirm this treatment outcome; in this abstract, a median survival of 13 months was reported.

Alberts et al (53) studied the combination of decortication, followed by systemic hemithoracic radiotherapy and systemic chemotherapy. Twenty-six patients were treated. The median survival was 10.9 months. Different combination of treatment modalities did not influence survival. (53)

Another study performed by Mattson et al, (54) with 100 patients included, showed a median survival of 8 months and a 2-year survival of 20%. Five different radiotherapy and chemotherapy regimens were used, but no statistical differences were seen between the groups. (54)

The combination of pleurectomy, external radiotherapy, and systemic chemotherapy was also studied in Memorial Sloan-Kettering Cancer Institute. (52,72) This multimodality treatment resulted in a median survival of 21s month for epithelial mesothelioma and 11 months for fibrosarcomatous mesothelioma. (72)

In a more recent study, Rusch et al (55) reported results of hemithoracic radiotherapy after complete resection in 61 patients. Adjuvant radiotherapy at a median of 54 Gy was well tolerated, except for one esophageal fistula. Only 13% patients had a local recurrence. Distant metastases were seen in 70% of the patients. The median survival was 17 months, and a 3-year survival of 27% is described. For stage I/II, the median survival was 34 months. Based on these results, the group of Rusch et al (55) adapted this treatment regimen as standard treatment for patients with limited pleural mesothelioma.

Surgery and Emphasis on Systemic Chemotherapy

Huncharek et al (57) studied the combination of surgery with postoperative systemic chemotherapy (Table 4). The combination of chemotherapy consisted of cisplatin and doxorubicin or cisplatin and mitomycin C. The median survival was 21 months with a 2-year survival of 23.9%. (54)

A less favorable outcome was found by Ceresoli et al. (43) In this small series (16 patients), the chemotherapy was mostly cisplatin, doxorubicin, or a combination of these agents. The median survival was 14 months. (43)

Hasturk et al (58) treated 20 patients with pleurectomy followed by systemic chemotherapy (cisplatin and mitomycin C) and immunotherapy ([alpha]-interferon). This resulted in a median survival of 12 months and a 2-year survival of 15%. The survival was calculated from the onset of chemotherapy. (58)

DaValle et al (56) reported a median survival of 17.5 months. Adjuvant therapy consisted of doxorubicin alone or in combination with other agents or irradiation. The reported survival was no better than that of the 13 patients not receiving adjunctive therapy. This study was not a randomized controlled one. (56)

Surgery and Emphasis on Intrapleural Chemotherapy

Intracavitary chemotherapy has the advantage of high local concentrations of the cytostatic drug while having limited systemic side effects. (73-75) Only direct cytotoxic agents appear rational. The pharmacokinetics of cisplatin and mitomycin are advantageous, but also show significant and sustained plasma levels. (74-76) One of the limiting factors is that the penetration depth of chemotherapy is limited to a few millimeters. (3) Therefore, intrapleural chemotherapy can only be profitable if it is preceded by optimal cytoreduction.

In a study performed by Lee et al (62) with intrapleural cisplatin and cytosine arabinoside after incomplete surgery (pleurectomy/decortication), the median survival was 11.5 months. Rusch et al, (60) Colleoni et al, (63) Sauter et al, (61) and Rice et al (59) studied the use of intrapleural chemotherapy after complete cytoreduction. All patients in these studies received adjuvant systemic chemotherapy. Rusch et al (60) studied the effect of instillation with cisplatin and mitomycin after pleurectomy or decortication. The median survival was 18 months, with a 2-year survival of 40%. The mortality was 3.7%, and significant morbidity was observed in 55%. Chemotherapy-related nephrotoxicity was seen in three patients (11%).a Recurrences were seen in 17 of 27 treated patients (63%). All recurrences, except one, were ipsilaterally localized. (51) Colleoni et al (63) applied cisplatin and cytarabine as intrapleural instillation after pleurectomy in 20 patients. One patient had a grade IV nephrotoxicity requiring dialysis. The overall median survival was 11.5 months; patients with minimal residual disease after pleurectomy had a median survival of 24.5 months,aa In the study of Sauter et al, (61) 13 patients received subtotal pleurectomy followed by intrapleural cisplatin and arabinosylcytosine, resulting in a median survival of 9 months with a 2-year survival of 25%. Rice et al (59) studied 19 patients with stage I MPM undergoing EPP or pleurectomy followed by postoperative intrapleural cisplatin and mitomycin. Grade I/II hematologic toxicity was seen in seven patients (58%). Mild ototoxicity was noticed in one patient. The mortality was 5%. Complications requiring reoperation developed in 16% of the patients. The median survival was 13 months. The site of recurrence was local (58%), distant (17%), or both local and distant (25%). (59)

Hyperthermia itself is cytotoxic; it enhances the cytotoxic effect of the cytostatic drugs, and it stimulates the penetration depth. (77-81) Carry et al (82) studied the addition of hyperthermia to surgery and intrapleural chemotherapy. Three patients with MPM stage I were included in this study. After pleurectomy, an intrapleural perfusion with mitomycin C was performed during 60 min. Because the risk of pulmonary edema is present at temperatures > 43C[degrees]C, the maximal pleural temperature was 42.6[degrees]C. (83) The technique was considered safe and feasible. No systemic toxic levels of mitomycin C were found. Two patients died after 4 months and 11 months, respectively, and one patient survived at least 22 months. (82) Yellin et al (84) treated seven patients with mesothelioma. A combination of surgery and intraoperative hyperthermic pleural perfusion with cisplatin over 60 min was used. The technique was feasible, easy to perform, and relatively safe. A median survival of 15 months was reported, with two patients surviving > 30 months. (84)

A multimodality therapy including surgery, pleural space perfusion with cisplatin, hyperthermia, and postoperative radiotherapy was studied by Ratto et al. (85) The duration of perfusion was 60 min in this study. Radiotherapy (55 Gy) was administered to chest wall incisions. Ten procedures were without any death or toxicity. Ratto et al (85) found higher systemic drug concentrations after pleurectomy/ decortication than after pleuropneumonectomy, indicating that the lung plays an important role in cisplatin absorption from the pleural space. Normothermic pleural space perfusion was performed in three patients. The local tissue/perfusate ratio of platinum concentrations tended to be higher after hyperthermic perfusion rather than normothermic perfusion. (85)

In the Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, we studied patients with MPM stage I treated with cytoreduction and intraoperative hyperthermic intrathoracic chemotherapy. (86) Cisplatin and doxorubicin were perfused over 90 min under mild hyperthermic conditions (40[degrees]C to 41[degrees]C). Doxorubicin was chosen because its enhanced activity under hyperthermic conditions; however, the penetration depth is limited. (87,88) Radiotherapy (8 Gy three times) on the thoracotomy scar and drainage tracts was administered to prevent scar recurrences. (4) The treatment was feasible but was accompanied by considerable toxicity. In a report of 11 patients, a median survival of 8 months was found. Disease recurred in three patients after 4 months, 6 months, and 7 months, respectively. The longest survivor without disease was 8 months. (86) An update of our results in 20 patients showed a median survival of 11 months (unpublished data).

Surgery and Emphasis on Intrapleural Photodynamic Therapy

Photodynamic therapy (PDT) has been considered a new mode of adjuvant treatment to sterilize the surgical field. After systemic administration of a photosensitizer, (tumor) cell kill can be achieved by illuminating the resection field with laser light. This principle was first tested by Takita and Dougherty, (89) who used a first-generation photosensitizer (Photofrin; Quadra Logic Technologies; Vancouver, BC). He treated 31 patients and reported a median survival of 12 months. The estimated median survival increased to 21 months when subdivided for stage I/II. Both pleurectomy and EPP were performed to achieve optimal cytoreduction. The mortality was 6.5%. Serious complications were observed in 48.3%, consisting of infection, bronchopleural fistula, cardiac arrhythmia, prolonged ventilatory support, chylothorax, hematothorax, and spontaneous rupture of the spleen. (89) A long-term report of the same institute including 40 patients revealed a median survival of 15 months. In stage I/II, the median survival was 36 months. (65)

An important randomized controlled study using PDT was performed by Pass et al. (64) Photofrin, a first-generation photosensitizer with a long illumination time, was used. Forty-eight patients underwent debulking to, at most, 5-mm residue. He found no survival benefit or improved local control for patients undergoing EPP pleurectomy combined with PDT. (64) The median survival was 14 months. The mortality and morbidity in this study were considerable: 2.1% and 20.8%, respectively. Complications such as death, bronchopleural fistula, esophageal perforation, and empyema are frequently seen when using PDT. (15,64)

Baas et al (90) studied intraoperative PDT after EPP in five patients using a second-generation photosensitizer (meta-tetrahydroxyphenylchlorin). The feasibility study was promising, but in the extended phase I/II study of 28 patients, the median survival was only 10 months. (66,90) In this study, three patients died in the perioperative period; one death was directly related to inappropriately delivered PDT, and two patients with advanced cases died as a result of cardiac complications. The considerable morbidity and mortality preclude this setup for widespread use. (66)

Escalating the light dose, improvement of light delivery, and addition of chemotherapy and radiotherapy are currently being investigated. Distant tumor spread is not prevented by this combined treatment modality. (91)

DISCUSSION

Prospects for the Future

Reviewing the literature on treatment of MPM is not encouraging. Not only has little progress has been made in the treatment of this disease, it is also clear that very few systematic attempts have been made to evaluate the effects of treatment strategies. Almost without exception, reports are retrospective, with poorly defined patient groups and large variations in treatment schedules. Most reported studies can at best be classified as phase I type feasibility studies. There are very few properly structured phase II studies and no phase III studies at all, in which a treatment schedule has been randomly compared to no treatment. In this era of evidence-based medicine, we can only conclude that no evidence exists of proven effectiveness of any treatment in MPM.

What lessons can be learned from the accumulated experience? The staging of MPM remains difficult by any standard. A preoperative CT scan and mediastinoscopy seem at present to be the minimum requirements for adequate staging. The distinction between stage I and higher stages is often possible. The distinction within stage I according to the IMIG staging system, which is meant to determine operability, is far more difficult. (24) Anyone engaged in surgery for MPM is impressed by the variation of growth characteristics in different patients. Sometimes, the tumor has a clear sharp margin and can easily be separated from neighboring structures; at other times, infiltrative growth with accompanying fibrosis is so dense that any attempt on removal is an illusion. In the present staging system, these characteristics are not fully represented, but determine to a large extent the completeness of any surgery, be it decortication or pleuropneumonectomy. It seems evident that only patients with stage I MPM are candidates who could benefit from aggressive locoregional therapies. However, it is clear that this has not been the case in most of the presently reviewed studies.

In this review, we could not find clear arguments to choose between decortication and pleuropneumonectomy as a first-choice surgical strategy. In many cases, decortication is not feasible because involvement of lung parenchyma. When technically possible, decortication seems to result in roughly the same survival as does pleuropneumonectomy (or no treatment?), but operative mortality is slightly decreased. In this review, we have focused on several multimodality treatments. Surgery combined with external radiotherapy included the whole hemithorax as radiation field in contrast to those in which only the surgical scars were radiated. In selected patients, a clear survival benefit is found; however, when critically analyzed, only 1 to 2% of all patients with mesothelioma could benefit of this treatment. (18,67) Although differences are limited, there remains an impression that survival in the series with external radiotherapy is somewhat longer than in the series not including hemithorax radiotherapy (approximately 20-month median survival in recent reports (32,55) vs approximately 15-month median survival in other combination therapies (43,56-66)). Side effects of radiotherapy on the liver and heart are mentioned but not quantified, especially not in the long term.

Autopsy studies of patients with MPM revealed that more than one half of the patients had disseminated MPM. (18) Therefore, systemic chemotherapy seems to be a prerequisite, but the survival of series with the combination of surgery with systemic chemotherapy appears very similar to the surgery-alone series. (43,56-58) The same is valid for the studies on the combination of surgery with intrapleural chemotherapy. (59-63) The intrapleural chemotherapy approach has probably not yet shown its full potential, as only few drugs (doxorubicin and cisplatin) have been studied, and dosage can probably still be increased. The combination of surgery with PDT has not shown a clear improvement of median survival until now. Furthermore, physical aspects like dosimetry of the light makes general application of this treatment difficult. PDT as part of a multimodality approach cannot be recommended at this stage. (64-66)

The fact remains that the large majority of patients with MPM die of locoregional failure despite aggressive locoregional therapy. This is especially true if recurrences in adjacent cavities (pericardium, contralateral pleura, and abdomen) are considered as regional failure, as we believe they should. The high locoregional failure could be explained by the relative insensibility of MPM to radiotherapy and chemotherapy. Intensifying the therapy is limited by the intolerance of adjacent vital structures (especially the lung). (34)

The conclusion of this review can only be that at this moment no therapy has been adequately shown to have any proven benefit in the treatment of MPM. At this moment, the combination of complete surgery, being decortication or pleuropneumonectomy, in combination with hemithorax radiotherapy seems promising only in selected patients. Intrapleural hyperthermic chemotherapy clearly needs a better-designed study. Future adjuvant therapies will also focus on gene therapy, small molecules (like tyrosine kinase inhibitors), and angiogenesis inhibitors. (15) For gene therapy, however, results have been disappointing given the remarkable results in animals. (92) Future studies would provide more useful information if they used a randomized phase II design, comparing the defined treatment with a no-treatment arm, especially if this would involve a quality-of-life assessment.

REFERENCES

(1) Moskal TL, Urschel JD, Anderson TM, et al. Malignant pleural mesothelioma: a problematic review. Surg Oncol 1998; 7:5-12

(2) Pass HI, Pogrebniak HW. Malignant pleural mesothelioma. Curr Probl Surg 1993; 30:921-1012

(3) Aisner J. Current approach to malignant mesothelioma of the pleura. Chest 1995; 107:332S-344S

(4) Boutin C, Rey F, Viallat JR. Prevention of malignant seeding after invasive diagnostic procedures in patients with pleural mesothelioma: a randomized trial of local radiotherapy. Chest 1995; 108:754-758

(5) Hillerdal G. Malignant mesothelioma 1982: review of 4710 published cases. Br J Dis Chest 1983; 77:321-343

(6) Janssen-Heijnen ML, Damhuis RA, Klinkhamer PJ, et al. Increased but low incidence and poor survival of malignant mesothelioma in the southeastern part of the Netherlands since 1970: a population-based study. Eur J Cancer Prev 1999; 8:311-314

(7) Muscat JE, Wynder EL. Cigarette smoking, asbestos exposure, and malignant mesothelioma. Cancer Res 1991; 51: 2263-2267

(8) Antman KH. Natural history and epidemiology of malignant mesothelioma. Chest 1993; 103:373S-376S

(9) Carbone M, Kratzke RA, Testa JR. The pathogenesis of mesothelioma. Semin Oncol 2002; 29:2-17

(10) Calavrezos A, Koschel G, Husselmann H, et al. Malignant mesothelioma of the pleura: a prospective therapeutic study of 132 patients from 1981-1985. Klin Wochenschr 1988; 66:607-613

(11) Ruffle P, Feld R, Minkin S, et al. Diffuse malignant mesothelioma of the pleura in Ontario and Quebec: a retrospective study of 332 patients. J Clin Oncol 1989; 7:1157-1168

(12) Baas P, Schouwink H, Zoetmulder FA. Malignant pleural mesothelioma. Ann Oncol 1998; 9:139-149

(13) Rusch VW. Clinical features and current treatment of diffuse malignant pleural mesothelioma. Lung Cancer 1995; 12(Suppl 2):S127-S146

(14) Ong ST, Vogelzang NJ. Chemotherapy in malignant pleural mesothelioma: a review. J Clin Oncol 1996; 14:1007-1017

(15) Sterman DH, Kaiser LR, Albelda SM. Advances in the treatment of malignant pleural mesothelioma. Chest 1999; 116:504-520

(16) Gordon W, Antman KH, Greenberger JS, et al. Radiation therapy in the management of patients with mesothelioma. Int J Radiat Oncol Biol Phys 1982; 8:19-25

(17) Bissett D, Macbeth FR, Cram I. The role of palliative radiotherapy in malignant mesothelioma. Clin Oncol (R Coll Radiol) 1991; 3:315-317

(18) Lee YC, Light RW, Musk AW. Management of malignant pleural mesothelioma: a critical review. Curr Opin Pulm Med 2000; 6:267-274

(19) Ho L, Sugarbaker DJ, Skarin AT. Malignant pleural mesothelioma. Cancer Treat Res 2001; 105:327-373

(20) Rusch VW. Pleurectomy/decortication and adjuvant therapy for malignant mesothelioma. Chest 1993; 103:382S-384S

(21) Rusch VW, Venkatraman E. The importance of surgical staging in the treatment of malignant pleural mesothelioma. Thorac Cardiovasc Surg 1996; 111:815-825

(22) Butchart EG, Ashcroft T, Barnsley WC, et al. Pleuropneumonectomy in the management of diffuse malignant mesothelioma of the pleura: experience with 29 patients. Thorax 1976; 31:15-24

(23) Sugarbaker DJ, Strauss GM, Lynch TJ, et al. Node status has prognostic significance in the multimodality therapy of diffuse, malignant mesothelioma. J Clin Oncol 1993; 11:1172-1178

(24) Rusch VW. A proposed new international TNM staging system for malignant pleural mesothelioma: The International Mesothelioma Interest Group. Chest 1995; 108:1122-1128

(25) Maasilta P, Vehmas T, Kivisaari L, et al. Correlations between findings at computed tomography (CT) and at thoracoscopy/ thoracotomy/autopsy in pleural mesothelioma. Eur Respir J 1991; 4:952-954

(26) Conlon KC, Rusch VW, Gillern S. Laparoscopy: an important tool in the staging of malignant pleural mesothelioma. Ann Surg Oncol 1996; 3:489-494

(27) Heelan RT, Rusch VW, Begg CB, et al. Staging of malignant pleural mesothelioma: comparison of CT and MR imaging. AJR Am J Roentgenol 1999; 172:1039-1047

(28) Rusch VW, Venkatraman ES. Important prognostic factors in patients with malignant pleural mesothelioma, managed surgically. Ann Thorac Surg 1999; 68:1799-1804

(29) Benard F, Sterman D, Smith RJ, et al. Metabolic imaging of malignant pleural mesothelioma with fluorodeoxyglucose positron emission tomography. Chest 1998; 114:713-722

(30) Patz EF Jr, Shaffer K, Piwnica-Worms DR, et al. Malignant pleural mesothelioma: value of CT and MR imaging in predicting resectability. AJR Am J Roentgenol 1992; 159:961-966

(31) Pass HI, Temeck BK, Kranda K, et al. Preoperative tumor volume is associated with outcome in malignant pleural mesothelioma. Thorac Cardiovasc Surg 1998; 115:310-317

(32) Sugarbaker DJ, Garcia JP, Richards WG, et al. Extrapleural pneumonectomy in the multimodality therapy of malignant pleural mesothelioma: results in 120 consecutive patients. Ann Surg 1996; 224:288-294

(33) Boutin C, Rey F, Gouvernet J, et al. Thoracoscopy in pleural malignant mesothelioma: a prospective study of 188 consecutive patients; part 2: prognosis and staging. Cancer 1993; 72:394-404

(34) Rusch VW. Pleurectomy/decortication in the setting of multimodality treatment for diffuse malignant pleural mesothelioma. Semin Thorac Cardiovasc Surg 1997; 9:367-372

(35) Soysal O, Karaoglanoglu N, Demiracan S, et al. Pleurectomy/ decortication for palliation in malignant pleural mesothelioma: results of surgery. Eur J Cardiothorac Surg 1997; 11:210-213

(36) Chahinian AP, Pajak TF, Holland JF, et al. Diffuse malignant mesothelioma: prospective evaluation of 69 patients. Ann Intern Med 1982; 96:746-755

(37) Brenner J, Sordillo PP, Magill GB, et al. Malignant mesothelioma of the pleura: review of 123 patients. Cancer 1982; 49:2431-2435

(38) Law MR, Gregor A, Hodson ME, et al. Malignant mesothelioma of the pleura: a study of 52 treated and 64 untreated patients. Thorax 1984; 39:255-259

(39) Chailleux E, Dabouis G, Pioche D, et al. Prognostic factors in diffuse malignant pleural mesothelioma: a study of 167 patients. Chest 1988; 93:159-162

(40) Brancatisano RP, Joseph MG, McCaughan BC. Pleurectomy for mesothelioma. Med J Aust 1991; 154:455-457,460

(41) Rusch VW, Piantadosi S, Holmes EC. The role of extrapleural pneumonectomy in malignant pleural mesothelioma: Lung Cancer Study Group trial. Thorac Cardiovasc Surg 1991; 102:1-9

(42) Allen KB, Faber LP, Warren WH. Malignant pleural mesothelioma: extrapleural pneumonectomy and pleurectomy. Chest Surg Clin N Am 1994; 4:113-126

(43) Ceresoli GL, Locati L, Ferreri AJ, et al. Therapeutic outcome according to histologic subtype in 121 patients with malignant pleural mesothelioma. Lung Cancer 2001; 34:279-287

(44) Hilaris BS, Nori D, Kwong E, et al. Pleurectomy and intraoperative brachytherapy and postoperative radiation in the treatment of malignant pleural mesothelioma. Int J Radiat Oncol Biol Phys 1984; 10:325-331

(45) Sugarbaker DJ, Mentzer SJ, DeCamp M, et al. Extrapleural pneumonectomy in the setting of a multimodality approach to malignant mesothelioma. Chest 1993; 103:377S-381S

(46) Sugarbaker DJ, Flores RM, Jaklitsch MT, et al. Resection margins, extrapleural nodal status, and cell type determine postoperative long-term survival in trimodality therapy of malignant pleural mesothelioma: results in 183 patients. Thorac Cardiovasc Surg 1999; 117:54-63

(47) Politi L, De Anna D, Navarra G, et al. Extrapleural pneumonectomies with pericardial and diaphragmatic plasty. Minerva Chir 2001; 56:251-255

(48) Harpole DH, Liptay MJ, DeCamp MM Jr, et al. Prospective analysis of pneumonectomy: risk factors for major morbidity and cardiac dysrhythmias. Ann Thorac Surg 1996; 61:977-982

(49) Worn H. [Chances and results of surgery of malignant mesothelioma of the pleura (author's translation).] Moglichkeiten und Ergebnisse der chirurgischen Behandlung des malignen Pleuramesothelioms. Thoraxchir Vask Chir 1974; 22:391-393

(50) Baldini EH, Recht A, Strauss GM, et al. Patterns of failure after trimodality therapy for malignant pleural mesothelioma. Ann Thorac Surg 1997; 63:334-338

(51) Rusch VW. Trials in malignant mesothelioma: LCSG 851 and 882. Chest 1994; 106:359S-362S

(52) McCormack PM, Nagasaki F, Hilaris BS, et al. Surgical treatment of pleura] mesothelioma. Thorac Cardiovasc Surg 1982; 84:834-842

(53) Alberts AS, Falkson G, Goedhals L, et al. Malignant pleural mesothelioma: a disease unaffected by current therapeutic maneuvers. J Clin Oncol 1988; 6:527-535

(54) Mattson K, Holsti LR, Tammilehto L, et al. Multimodality treatment programs for malignant pleural mesothelioma using high-dose hemithorax irradiation. Int J Radiat Oncol Biol Phys 1992; 24:643-650

(55) Rusch VW, Rosenzweig K, Venkatraman E, et al. A phase II trial of surgical resection and adjuvant high-dose hemithoracic radiation for malignant pleural mesothelioma. Thorac Cardiovasc Surg 2001; 122:788-795

(56) DaValle MJ, Faber LP, Kittle CF, et al. Extrapleural pneumonectomy for diffuse, malignant mesothelioma. Ann Thorac Surg 1986; 42:612-618

(57) Huncharek M, Kelsey K, Mark EJ, et al. Treatment and survival in diffuse malignant pleural mesothelioma: a study of 83 cases from the Massachusetts General Hospital. Anticancer Res 1996; 16:1265-1268

(58) Hasturk S, Tastepe I, Unlu M, et al. Combined chemotherapy in pleurectomized malignant pleural mesothelioma patients. J Chemother 1996; 8:159-164

(59) Rice TW, Adelstein DJ, Kirby TJ, et al. Aggressive multimodality therapy for malignant pleural mesothelioma. Ann Thorac Surg 1994; 58:24-29

(60) Rusch V, Saltz L, Venkatraman E, et al. A phase II trial of pleurectomy/decortication followed by intrapleural and systemic chemotherapy for malignant pleural mesothelioma. J Clin Oncol 1994; 12:1156-1163

(61) Sauter ER, Langer C, Coia LR, et al. Optimal management of malignant mesothelioma after subtotal pleurectomy: revisiting the role of intrapleural chemotherapy and postoperative radiation. J Surg Oncol 1995; 60:100-105

(62) Lee JD, Perez S, Wang HJ, et al. Intrapleural chemotherapy for patients with incompletely resected malignant mesothelioma: the UCLA experience. J Surg Oncol 1995; 60:262-267

(63) Colleoni M, Sartori F, Calabro F, et al. Surgery followed by intracavitary plus systemic chemotherapy in malignant pleural mesothelioma. Tumori 1996; 82:53-56

(64) Pass HI, Temeck BK, Kranda K, et al. Phase III randomized trial of surgery with or without intraoperative photodynamic therapy and postoperative immunochemotherapy for malignant pleural mesothelioma. Ann Surg Oncol 1997; 4:628-633

(65) Moskal TL, Dougherty TJ, Urschel JD, et al. Operation and photodynamic therapy for pleural mesothelioma: 6-year follow-up. Ann Thorac Surg 1998; 66:1128-1133

(66) Schouwink H, Rutgers ET, van der Sijp J, et al. Intraoperative photodynamic therapy after pleuropneumonectomy in patients with malignant pleural mesothelioma: dose finding and toxicity results. Chest 2001; 120:1167-1174

(67) Sugarbaker DJ, Jaklitsch MT, Liptay MJ. Mesothelioma and radical multimodality therapy: who benefits? Chest 1995; 107:345S-350S

(68) Rubin P, Constine LS, Nelson DF. Late effects of cancer treatment: radiation and drug toxicity. In: Perez CA, Brady LW, eds. Principles and practice of radiation oncology. Philadelphia, PA: J.B. Lippincott, 1992;124-161

(69) Maasilta P. Deterioration in lung function following hemithorax irradiation for pleural mesothelioma. Int J Radiat Oncol Biol Phys 1991; 20:433-438

(70) Grondin SC, Sugarbaker DJ. Pleuropneumonectomy in the treatment of malignant pleural mesothelioma. Chest 1999; 116:450S-454S

(71) Mychalczak BR, Nori D, Armstrong JG, et al. Results of treatment of malignant pleural mesothelioma with surgery, brachytherapy, and external beam radiation [abstract]. Endocurie Hypertherm Oncol 1989; 5:245

(72) Wanebo HJ, Martini N, Melamed MR, et al. Pleural mesothelioma. Cancer 1976; 38:2481-2488

(73) Markman M. Intraperitoneal chemotherapy. Semin Oncol 1991; 18:248-254

(74) Bogliolo GV, Lerza R, Bottino GB, et al. Regional pharmacokinetic selectivity of intrapleural cisplatin. Eur J Cancer 1991; 27:839-842

(75) Rusch VW, Niedzwiecki D, Tao Y, et al. Intrapleural cisplatin and mitomycin for malignant mesothelioma following pleurectomy: pharmacokinetic studies. J Clin Oncol 1992; 10: 1001-1006

(76) Lerza R, Esposito M, Vannozzi M, et al. High doses of intrapleural cisplatin in a case of malignant pleural mesothelioma: clinical observations and pharmacokinetic analyses. Cancer 1994; 73:79-84

(77) van de Vaart PJ, van der Vange N, Zoetmulder FA, et al. Intraperitoneal cisplatin with regional hyperthermia in advanced ovarian cancer: pharmacokinetics and cisplatin-DNA adduct formation in patients and ovarian cancer cell lines. Eur J Cancer 1998; 34:148-154

(78) Storm FK. Clinical hyperthermia and chemotherapy. Radiol Clin North Am 1989; 27:621-627

(79) Los G, van Vugt MJ, Pinedo HM. Response of peritoneal solid tumours after intraperitoneal chemohyperthermia treatment with cisplatin or carboplatin. Br J Cancer 1994; 69:235-241

(80) Ohtsubo T, Saito H, Tanaka N, et al. In vitro effect of hyperthermia on chemoenhancement and uptake of cisplatin in human pharyngeal carcinoma KB cells. Chemotherapy 1997; 43:43-50

(81) Los G, Sminia P, Wondergem J, et al. Optimisation of intraperitoneal cisplatin therapy with regional hyperthermia in rats. Eur J Cancer 1991; 27:472-477

(82) Carry PY, Brachet A, Gilly FN, et al. A new device for the treatment of pleural malignancies: intrapleural chemohyperthermia preliminary report. Oncology 1993; 50:348-352

(83) Mustafa KY, Selig WM, Burhop KE, et al. Hyperthermia-induced pulmonary edema. J Appl Physiol 1986; 60:1980-1985

(84) Yellin A, Simansky DA, Paley M, et al. Hyperthermic pleural perfusion with cisplatin. Cancer 2001; 92:2197-2203

(85) Ratto GB, Civalleri D, Esposito M, et al. Pleural space perfusion with cisplatin in the multimodality treatment of malignant mesothelioma: a feasibility and pharmacokinetic study. Thorac Cardiovasc Surg 1999; 117:759-765

(86) de Bree E, van Ruth S, Baas P, et al. Cytoreductive surgery and intraoperative hyperthermic intrathoracic chemotherapy in patients with malignant pleural mesothelioma or pleural metastases of thymoma. Chest 2002; 121:480-487

(87) Hahn GM, Braun J, Har-Kedar I. Thermochemotherapy: synergism between hyperthermia (42-43 degrees) and adriamycin (of bleomycin) in mammalian cell inactivation. Proc Natl Acad Sci U S A 1975; 72:937-940

(88) Ozols RF, Locker GY, Doroshow JH, et al. Pharmacokinetics of adriamycin and tissue penetration in murine ovarian cancer. Cancer Res 1979; 39:3209-3214

(89) Takita H, Dougherty TJ. Intracavitary photodynamic therapy for malignant pleural mesothelioma. Semin Surg Oncol 1995; 11:368-371

(90) Baas P, Murrer L, Zoetmulder FA, et al. Photodynamic therapy as adjuvant therapy in surgically treated pleural malignancies. Br J Cancer 1997; 76:819-826

(91) Ris HB, Altermatt HJ, Nachbur B, et al. Intraoperative photodynamic therapy with m-tetrahydroxyphenylchlorin for chest malignancies. Lasers Surg Med 1996; 18:39-45

(92) Sterman DH, Treat J, Litzky LA, et al. Adenovirus-mediated herpes simplex virus thymidine kinase/ganciclovir gene therapy in patients with localized malignancy: results of a phase I clinical trial in malignant mesothelioma. Hum Gene Ther 1998; 9:1083-1092

* From the Department of Surgical Oncology (Drs. van Ruth and Zoetmulder) and Thoracic Oncology (Dr. Baas), The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands.

Manuscript received March 13, 2002; revision accepted June 13, 2002.

Correspondence to: Serge van Ruth, MD, Department of Surgical Oncology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; e-mail: s.v.ruth@nki.nl

COPYRIGHT 2003 American College of Chest Physicians
COPYRIGHT 2003 Gale Group

Return to Mesothelioma
Home Contact Resources Exchange Links ebay