The Immunogenic Effect of Local Radiation Therapy … · Luis Alberto de la Maza Borja ... was to...

164
The Immunogenic Effect of Local Radiation Therapy in a Murine Model of Mesothelioma by Luis Alberto de la Maza Borja A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Luis Alberto de la Maza Borja 2015

Transcript of The Immunogenic Effect of Local Radiation Therapy … · Luis Alberto de la Maza Borja ... was to...

The Immunogenic Effect of Local Radiation Therapy in a Murine Model of Mesothelioma

by

Luis Alberto de la Maza Borja

A thesis submitted in conformity with the requirements for the degree of Master of Science

Institute of Medical Science University of Toronto

© Copyright by Luis Alberto de la Maza Borja 2015

ii

The Immunogenic Effect of Local Radiation Therapy in a Murine

Model of Mesothelioma

Luis A. de la Maza Borja

Master of Science

Institute of Medical Science

University of Toronto

2015

Abstract

Malignant pleural mesothelioma (MPM) is a rare cancer that arises from the mesothelial surfaces

of the pleural cavity. It is associated with asbestos inhalation, with 70% of cases being associated

with documented asbestos exposure. MPM has a poor prognosis and the outlook has not been

improved by newer therapeutic interventions. A new approach developed in our lab, focusing on

Surgery for Mesothelioma After Radiation Therapy (SMART) showed promising results in a

phase I/II clinical trial. We believe that radiation is important to achieving activation of the

immune system and may contribute to the benefits observed in patients. The goal of this project

was to develop a mouse model to analyze the immunogenic effect of Local Radiation Therapy

(LRT) and its impact on immune cell recruitment and activation in the context of MPM. Results

from these studies may have clinical implication for the treatment of MPM, where combination

of LRT and other treatments such as immunotherapy may prove useful.

iii

Acknowledgments

I would like to thank my supervisor Dr. Marc de Perrot, the members of my lab, Licun Wu,

Yidan Zhao and Hannah Yun. Many thanks to my friend and bro Matthew Wu, together we

learned, grew and overcome difficulties in the lab and in our life projects. Also I would like to

thank the members of my PAC committee, Dr Pam Ohashi, Dr. Mark Cattral, Dr. Andrea

McCart and Dr. John Cho. Without the help and support of Carolyn, Ingrid, Corrina and Marlene

the meetings with my advisors would not have been possible. The support of my family many

kilometers away was instrumental in the completion of this work, specially my parents who

always believed in me and were curious about my work, Ana Maria G Borja Cano and Carlos A.

de la Maza Gonzalez. My brothers and sister who inspired me to keep working and supported me

through difficult times, Laura Patricia, Carlos Alejandro and Jose Eduardo. My uncle and aunt

Magda and Gerardo de la Maza Gonzales, without their support this would not have been

possible. Most importantly, my best friend, colleague and wife, Marisol Davila Foyo, who spent

many hours deliberating about my work, preparing assignments and listening to my

presentations, big thanks to you my love.

iv

Contributions

This work and my Master’s degree was supported partly by CONACyT and the MARF grant

(Mesothelioma Applied Research Foundation) and by the Princess Margaret Hospital

Foundation. Matthew Wu performed immunofluorescence staining of tumor samples.

v

Table of Contents

Contents

Contributions .................................................................................................................................. iv

Table of Contents ............................................................................................................................ v

List of Tables ................................................................................................................................. ix

List of Figures ................................................................................................................................. x

List of Abbreviations ..................................................................................................................... xi

Chapter 1 ......................................................................................................................................... 1

1 General Background ................................................................................................................... 1

1.1 Malignant Pleural Mesothelioma ........................................................................................ 1

1.1.1 Historical Background ............................................................................................ 1

1.1.2 Epidemiology .......................................................................................................... 2

1.1.3 Etiology ................................................................................................................... 4

1.1.4 Pathogenesis ............................................................................................................ 6

1.1.5 Clinical Presentation ............................................................................................... 8

1.1.6 Diagnosis ................................................................................................................. 9

1.1.7 Current Management ............................................................................................ 11

1.1.8 Prospective treatment Options for MPM .............................................................. 18

1.2 Tumor Immunity ............................................................................................................... 26

1.2.1 The immune response ........................................................................................... 26

1.2.2 Adaptive immunity ............................................................................................... 27

1.2.3 The T cell immune response ................................................................................. 28

1.2.4 Tumor Escape ....................................................................................................... 30

1.3 Radiation Therapy of tumors and the Immune System .................................................... 32

1.3.1 Brief history .......................................................................................................... 32

vi

1.3.2 Radiation and its interaction with matter .............................................................. 32

1.3.3 Factors affecting the cellular response to radiation .............................................. 34

1.3.4 Cellular Sensing and Response to Radiation ........................................................ 34

1.3.5 Cell death response ............................................................................................... 36

1.3.6 Immunogenic Cell Death ...................................................................................... 37

1.3.7 Tumor microenvironment ..................................................................................... 39

1.4 The immune response to surgery ...................................................................................... 42

1.4.1 The Surgical Stress response ................................................................................ 42

1.4.2 Post-surgical cytokine cascades ............................................................................ 44

1.4.3 Postoperative tumor progression ........................................................................... 46

1.5 Summary ........................................................................................................................... 46

2 Hypothesis and Aims ............................................................................................................... 49

3 Materials and Methods ............................................................................................................. 51

3.1 Murine Cell lines ............................................................................................................... 51

3.2 Mice .................................................................................................................................. 52

3.3 In vivo Tumor Growth experiments ................................................................................. 52

3.4 Local Radiation Therapy ................................................................................................... 52

3.5 Surgical Resection of Subcutaneous Tumors ................................................................... 53

3.6 Combination therapy with LRT and Surgery .................................................................... 55

3.7 In vivo depletion of CD4+ and CD8+ specific T cells ..................................................... 55

3.8 Anti-CTLA-4 therapy ....................................................................................................... 56

3.9 Blood Collection ............................................................................................................... 56

3.10 Tumor Digestion ............................................................................................................... 56

3.11 Isolation of Lymphocytes from Spleens and Lymph Nodes ............................................. 57

3.12 Flow Cytometry ................................................................................................................ 57

3.13 Immunofluorescence ......................................................................................................... 58

vii

3.14 Immunohistochemistry ..................................................................................................... 58

3.15 Ovalbumin ELISA ............................................................................................................ 59

3.16 Statistical Analysis ............................................................................................................ 60

4 Results ...................................................................................................................................... 61

4.1 Development of a Mouse Model of Malignant Mesothelioma ......................................... 61

4.1.1 Local Radiation Therapy, Right Flank Model ...................................................... 61

4.1.2 Combination therapy with LRT and Surgery ........................................................ 65

4.2 T cells infiltrate tumors after LRT .................................................................................... 68

4.2.1 Tumor infiltrating CD8+ T Cells in Untreated and Radiated Mouse Tumor

Tissue .................................................................................................................... 68

4.2.2 A Large Proportion of Tumor Infiltrating Lymphocytes are OVA-specific ......... 71

4.2.3 Expression of 4-1BB and PD-1 by Tumor Infiltrating Cells ................................ 73

4.2.4 Depletion of CD4+ T cells and CD8+ T cells partially abrogates the effect of

LRT on tumor growth ........................................................................................... 74

4.3 Immunological Protective Memory After LRT and Surgery ............................................ 77

4.3.1 Role of T cell on the protection against rechallenge ............................................. 79

4.4 CTLA-4 blockade improves the beneficial effect of LRT on tumors ............................... 82

5 Discussion ................................................................................................................................ 84

5.1 Development of a mouse model of MPM treated with LRT followed by surgery ........... 85

5.2 T cell tumor infiltration ..................................................................................................... 88

5.2.1 Upregulation of the activation marker 4-1BB and decrease in the exhaustion

marker PD-1 .......................................................................................................... 90

5.2.2 Depletion of lymphocytes partially abrogated the effect of radiation .................. 91

5.3 Protective memory response after LRT and Surgery ........................................................ 92

5.3.1 Depletion and rechallenge ..................................................................................... 93

5.4 CTLA-4 blockade synergized with LRT .......................................................................... 94

6 Conclusions .............................................................................................................................. 96

viii

7 Limitations ............................................................................................................................. 100

Future Directions ........................................................................................................................ 102

8 References .............................................................................................................................. 105

ix

List of Tables

Table 1 Worldwide Trends in the Epidemiologic Features of Malignant Mesothelioma

Table 2. Diagnosis of Mesothelioma

x

List of Figures

Figure 1. A possible mechanism for asbestos induced oncogenesis.. ............................................. 7

Figure 2. SMART Study schema. RT, radiotherapy. .................................................................... 17

Figure 3 Partial resection of a subcutaneous tumor with blunt dissection.. .................................. 54

Figure 4. Schematic of radiation treatment in tumor bearing mice. ............................................. 62

Figure 5 Increasing doses of Local Radiation Therapy and its effect on tumor growth.. ............. 64

Figure 6 Schematic of LRT and Surgery in AB12 tumor bearing mice.. ..................................... 66

Figure 7. The effect of combination therapy with LRT and Surgery.. ......................................... 67

Figure 8. CD3+CD8+ cell tumor infiltration after LRT compared to untreated tumors.. ............ 70

Figure 9. CD8+ lymphocytes infiltrating AE17-OVA tumor are OVA specific.. ........................ 72

Figure 10. 4-1BB and PD-1 expression tetramer specific CD8+ T cells after LRT. .................... 74

Figure 11. LRT and CD4+ CD8+ T cell depletion. ...................................................................... 76

Figure 12. AE17 OVA Rechallenge 90 days after treatment ........................................................ 78

Figure 13. CD4+ and CD8+ T cells role during rechallenge.. ...................................................... 81

Figure 14. Combination therapy with LRT and CTLA-4 shows a synergistic effect on tumor

growth. .......................................................................................................................................... 83

xi

List of Abbreviations

Ab Antibody

APC Antigen Presenting Cell

ATP Adenosine Triphosphate

BSA Bovine Serum Albumin

CRT Calreticulin

CTCF Corrected Total Cell Fluorescence

CTLA-4 Cytotoxic T-Lymphocyte-Associated Antigen 4

DAMP Danger Associated Molecular Pattern

DC Dendritic Cell

EGF Epidermal Growth Factor

ELISA Enzyme-Linked Immunosorbent Assay

EPD Extended Pleurectomy-Decortication

EPP Extrapleural Pneumonectomy

ERK Extracellular Signal-Related Kinase

FBS Fetal Bovine Serum

HMGB-1 High Mobility Group Box 1

ICAM Intracellular Adhesion Molecule

ICD Immunogenic Cell Death

IL Interleukin

LFA Lymphocyte Function Associated Antigen

LRT Local Radiation Therapy

mAb Monoclonal Antibody

Mac-1 Macrophage Receptor 1

MHC Major Histocompatibility

xii

MPM Malignant Pleural Mesothelioma

NF-κB Nuclear Factor Kappa Light Chain Enhancer of Activated B Cells

NK Natural Killer

NLRP3 NOD-like Receptor Family Pyrin Domain Containing 3

PD-1 Programmed Death-1

PD-L1 Programmed Death Ligand 1

PAMP Pathogen Associated Molecular Pattern

PRR Pathogen Recognition Receptor

RAGE Receptor for Advanced Glycation End Products

ROS Reactive Oxygen Species

SMART Surgery for Mesothelioma After Radiation Therapy

SV40 Simian Virus 40

TCR T Cell Receptor

TLR Toll-Like Receptor

TNF Tumor Necrosis Factor

VCAM Vascular Cell Adhesion Molecule

VLA Very Late Antigen

WT-1 Wilms Tumor Protein

1

Chapter 1

1 General Background

1.1 Malignant Pleural Mesothelioma

Mesothelioma is a rare and insidious malignancy with dismal prognosis. It arises from the

mesothelial surfaces of the pleural cavity, peritoneum, tunica vaginalis or pericardium. Pleural

mesothelioma is the most common type, accounting for about 70% of all malignant

mesothelioma cases (Yang, Testa, and Carbone 2008). The main risk factor associated with

development of MPM is chronic inhalational exposure to asbestos (Fuhrer and Lazarus 2011).

MPM is characterized by insidious growth and clinical presentation at an advanced stage of

disease. Currently, even with aggressive multimodality interventions including invasive surgery,

prognosis remains poor (Raja, Murthy, and Mason 2011).

1.1.1 Historical Background

The story of the discovery of mesothelioma and its causation by asbestos is long and complex.

The earliest mention of a possible tumor of the chest wall, likely mesothelioma, was by Joseph

Lieutaud in 1767. There were other mentions of patients with pleural tumors after this, but it was

only in 1870 that Wagner published the first pathological description of a primary malignancy of

2

the pleura, which he called endothelioma. By 1920, Du Bray and Rosson suggested that the term

endothelioma was not appropriate and proposed the term mesothelioma of the pleura (Hsu 2006).

In 1943, in Germany Dr. H.W. Wedler reported the first case of diffuse mesothelioma in a

patient with asbestosis, but the link between asbestos and MPM was not discovered until 1960 in

South Africa. In 1960, Wagner et al. reported a MPM epidemic among asbestos miners in the

North West of Cape Province. This was the first convincing link between asbestos exposure and

MPM (Wagner, Sleggs, and Marchand 1960).

1.1.2 Epidemiology

The worldwide incidence of Malignant Pleural Mesothelioma (MPM) is approximately 0.9 case

per 100,000 persons (Driece et al. 2010). The rate is variable between countries partly because of

asbestos exposure. In North America the incidence is about 2000-3000 new cases per year

(Yang, Testa, and Carbone 2008). In the United States (Bruce W S Robinson and Lake 2005) it

is possible that the disease has already reached its peak. Peak incidence is predicted to occur

between 2015 and 2020 in Canada (Bruce W S Robinson and Lake 2005; Cree et al. 2009),

Europe (Peto et al. 1999; Marinaccio et al. 2005; Hodgson et al. 2005) and Australia (Bruce W S

Robinson and Lake 2005; B. M. Robinson 2012; Clements et al. 2007). Moreover, In Japan (B.

M. Robinson 2012) and other non-western countries where asbestos regulation occurred later, the

peak in mesothelioma incidence will be delayed as well (Table 1).

3

Table 1 Worldwide Trends in the Epidemiologic Features of Malignant Mesothelioma

Country or Region Incidence (cases/million

population)(Bruce W S Robinson

and Lake 2005)

Predicted Peak Year

Canada 15 2015-2020 (Cree et al.

2009)

Italy 24 2015-2020 (Marinaccio et

al. 2005)

United Kingdom 29 2015-2020 (Hodgson et al.

2005)

Japan 8 2027 (B. M. Robinson

2012)

Australia 29 2015-2021 (B. M.

Robinson 2012; Clements

et al. 2007)

About 2 to 10% of individuals exposed to asbestos develop MPM. On the other hand, up to 80%

of MPM patients have a history of asbestos exposure (Yang, Testa, and Carbone 2008); (M

Carbone et al. 2003). MPM is more common in men than women, with a male to female ratio of

3:1 (Reid et al. 2005), or males being 68 to 79% of all cases (Ruffie et al. 1989; Adams et al.

1986; Antman et al. 1988; Brenner et al. 1982; Ratzer, Pool, and Melamed 1967). This greater

incidence in males is secondary to occupational exposure in men (Fuhrer and Lazarus 2011).

MPM is usually unilateral (90%) and commonly in the right thorax (60%) due to asbestos

concentrating more readily in the right lung (Cugell and Kamp 2004). The peak incidence is in

4

between the 6th and 8th decade of life and in up to 80% of patients a past asbestos exposure can

be identified (Ismail-Khan et al. 2006).

1.1.3 Etiology

Chronic asbestos exposures is the most important risk factor associated with subsequent

development of pleural mesothelioma (Ismail-Khan et al. 2006; Raja, Murthy, and Mason 2011;

Suzuki, Yuen, and Ashley 2005; Yang, Testa, and Carbone 2008). The word asbestos comes

from the ancient Greek ἄσβεστος, meaning "inextinguishable" (Yang, Testa, and Carbone 2008).

There are six minerals defined by the United States Environmental Protection Agency as

“asbestos”. The two major forms of asbestos are the serpentine and amphibole. Serpentine or

white asbestos are feathery fibers and the only member of this class is chrysotile. Amphibole

asbestos is defined by long and thin fibers and include crocidolite (blue asbestos) (Leigh and

Robinson 2002; Hodgson and Darnton 2000; Bruce W S Robinson and Lake 2005). The

association of amphibole asbestos exposure and MPM is well accepted but whether chrysotile

fibers carries less risk of mesothelioma is still debated (Suzuki, Yuen, and Ashley 2005;

Yarborough 2006; Powers and Carbone 2002; Hodgson and Darnton 2000).

SV40 (Simian Virus 40), a DNA virus, has been implicated as a cofactor in the causation of

MPM (M Carbone et al. 1994). This virus blocks tumor-suppressor genes and is a potent

oncogenic virus in human and rodent cells; SV40 DNA has been found in MPM and atypical

mesothelial proliferations (Shivapurkar et al. 2002). SV40 is believed to have contaminated the

Salk polio vaccine that was used from 1955 to 1963 in the US. However, epidemiologic data is

not consistent with an etiologic effect of exposure to SV40-contaminated polio virus and the

5

development of cancer (Bocchetta and Carbone 2005; Fisher, Weber, and Carbone; Heinonen et

al. 1973; Innis 1968; Strickler et al. 1998). Subsequently, SV40 was considered harmless to

humans for many years. Finally, during the past decade interest in the association of SV40 and

human cancer has resurfaced and now the experimental data strongly associates SV40 with

human tumors, and more specifically with MPM (M Carbone et al. 2003; Bocchetta et al. 2000;

Jasani et al. 2001; Gazdar, Butel, and Carbone 2002).

Genetics may also play a role in MPM as demonstrated in some Cappadocian villages in Turkey

where an MPM epidemic caused up to 50% of all deaths (Michele Carbone et al. 2007). In these

villages, MPM was prevalent in some families but not others, even though all houses contained

similar levels of erionite. Erionite is a different type of mineral fiber and is one of the most

potent inducers of MPM in animal studies (Hill, Edwards, and Carthew 1990). MPM appeared to

be inherited in an autosomal dominant pattern according to pedigree studies. The result of

mineralogical studies and pedigree analysis suggested that MPM in Cappadocia is caused by

erionite exposure in a genetically predisposed population (gene and environment) (Yang, Testa,

and Carbone 2008). Recent work done by Dr. Carbone has led to the identification of BAP1 as

the gene mutated and associated with high rates of mesothelioma in 2 clusters of families in

United States and possibly in Cappadocia (Michele Carbone et al. 2013).

Radiation has also been linked with MPM development in animal studies (Sanders and Jackson

1972). Furthermore, multiple case reports have documented MPM in patients who received

radiation therapy for other malignancies decades before, with an average interval of 21 years

(Yang, Testa, and Carbone 2008; Amin, Mason, and Rowe 2001; Travis et al. 2005).

6

1.1.4 Pathogenesis

The development of MPM and asbestos pathogenicity are still not fully understood (Michele

Carbone, Kratzke, and Testa 2002). It is thought that asbestos is inhaled and fibers reach the

alveoli. Fibers are not easily removed and eventually translocate to the pleura via the lymphatics

or by direct extension (Powers and Carbone 2002). Asbestos fibers in the pleural space cause

persistent inflammation and secretion of cytokines, recruitment of macrophages and neutrophils

(Choe et al. 1997). Pleural mesothelial cells (PMC) secretes monocyte chemoattractant protein-1

(MCP-1) in response to asbestos. MCP-1 attracts monocytes that in turn differentiate into

macrophages and causes accumulation of macrophages within the pleural space (Tanaka et al.

2000). Upon differentiation into macrophages these cells phagocytose, and together with pleural

mesothelial cells release tumor necrosis factor alpha (TNF-α) and IL-1beta (IL-1β) (Y. Zhang et

al. 1993; Wang et al. 2004). Asbestos, also induces the expression of the TNF- α receptor 1

(TNF-R1) on pleural mesothelial cells. Activation of the TNF- α receptor in PMC consequently

activates NF-κB signaling that promotes survival and division of these cells (Yang et al. 2006).

Concurrently, the chronic state of inflammation upregulates other growth factors, including

platelet derived growth factor (PDGF), insulin-like growth factor (IGF), fibroblast growth factor

(FGF), and tumor growth factor (TGF). These factors appear to stimulate mesothelioma cell

proliferation and angiogenesis (Fuhrer and Lazarus 2011; Liu and Klominek 2004). The damage

and repair cycle contributes to DNA damage and subsequent transformation of PMC into

cancerous cells.

7

Figure 1. A possible mechanism for asbestos induced oncogenesis. TNF-α inhibits asbestos

cytotoxicity via an NF-κβ dependent pathway. Reproduced With kind permission from Springer

Science+Business Media: Current Treatment Options in Oncology, Mesothelioma Epidemiology, Carcinogenesis, and

Pathogenesis, volume 9, 2008, p. 10, Yang H; Testa JR and Carbone M, figure 1.

Asbestos fibers have also been reported to disturb mitosis by damaging the mitotic spindle of

cells, resulting in aneuploidy. The asbestos fibers may also cause DNA strand breaks secondary

to the release of iron-catalyzed free radicals (Kamp et al. 1995). In addition, asbestos causes the

release of reactive oxygen species (ROS) and reactive nitrogen species (RNS) which are

8

genotoxic and lead to a broad spectrum of mutations (Xu et al. 2002). Another mechanism by

which asbestos affects the pleura is induced phosphorylation of mitogen-activated protein (MAP)

kinases and extracellular signal-regulated kinases (ERK) 1 and 2. The phosphorylation of these

proteins increases expression of oncogenes such as activator protein (AP)-1 and subsequent

increased mitosis of PMC (Ramos-Nino, Timblin, and Mossman 2002).

In summary, asbestos promotes a chronic inflammatory environment, repeated cell injury and

DNA damage together with expression of cytokines and chemokines promoting proliferation,

survival and the activation of oncogenes. This may lead to PMC accumulating DNA damage and

subsequently development of MPM.

1.1.5 Clinical Presentation

Patients with MPM typically present with progressive dyspnea, chest wall pain and pleural

effusion. Dyspnea develops secondary to large pleural effusions and chest wall pain is a

consequence of chest wall infiltration. Mesothelioma is suspected in any patient with

unexplained pleural effusion and chest wall pain (Bruce W S Robinson, Musk, and Lake 2005).

Other symptoms include cough, fatigue and a mass on the chest wall. Constitutional symptoms

like weight loss, fever, night sweats and cachexia may be present in late stages of MPM.

Presentation of these symptoms at diagnosis of MPM is associated with poor prognosis (Bruce

W S Robinson, Musk, and Lake 2005).

On physical exam, the most common findings are those of pleural effusion, that is, dullness to

percussion on the affected side, decreased breath sounds and a lack of egophony. Clubbing

9

occurs in in less than 1 percent of cases (Raja, Murthy, and Mason 2011; Fuhrer and Lazarus

2011).

There are no specific laboratory findings diagnostic of MPM but eosinophilia, thrombocytosis,

and anemia of chronic diseases may be present (Ruffie et al. 1989).

1.1.6 Diagnosis

Physical exam and chest X-ray show pleural effusion in most cases (80% to 95%). Imaging

studies and biopsy are needed for definitive diagnosis. Computed chest tomography (CT) with

contrast is sensitive and is the one of the most frequent technique used to evaluate patients with

MPM. CT is sensitive to detect pleural effusion, pleural masses and lymph nodes in the hilum

and mediastinum. Magnetic resonance imaging (MRI) of the chest with contrast can provide

more information on chest wall and diaphragm invasion that may be important when considering

curative surgery. Positron emission tomography (PET) offers additional information when

assessing patients, occasionally demonstrating metastasis to contralateral thorax or extrathoracic

lymph nodes (Marom et al. 2002).

Pleural fluid cytology has a low sensitivity for diagnosis of MPM, yielding a positive diagnosis

in 33 to 84% of patients (Whitaker 2000). Blind core biopsy only modestly increases the

sensitivity. A CT-guided core needle biopsy is 87% sensitive depending on the bulkiness of the

disease and video-assisted thoracoscopy (VATS) directed pleural biopsy may increase diagnostic

accuracy to 95% to 98%. However, this procedure increases the possibility of seeding tumor

cells along the surgical incision. Some authors report tumor growth in the chest wall after VATS

10

in up to 20% of patients (Ismail-Khan et al. 2006; Bruce W S Robinson, Musk, and Lake 2005;

Nguyen et al. 1999).

Immuno-histochemical staining is usually needed for the definitive diagnosis of MPM. The most

common diagnostic problem is distinguishing mesothelioma from adenocarcinoma (Tang et al.

2001). MPM is typically positive for calretinin (88%), vimentin (50%), Wilm’s tumor 1 (WT1)

and epithelial membrane antigen (EMA, 85%) (Bruce W S Robinson, Musk, and Lake 2005).

Adenocarcinoma usually lacks these markers and is positive for carcinoembryonic antigen

(84%), CD15 (77%) and Ber-EP-4 (82%) (Martensson 1990). Histological diagnosis is also

useful to determine the subtype. Epithelial mesothelioma which has the best prognosis is the

most common and is present in 60 to 70% of cases. Sarcomatoid, characterized by spindle-

shaped cells is more aggressive and is present in 10-15% of cases. Biphasic, a combination of

epithelial and sarcomatoid, is seen in 15-30% of patients (Tischoff et al. 2011).

Despite many diagnostic options, it is common that a definitive diagnosis of MPM is delayed due

to low clinical suspicion for the disease.

Table 2. Diagnosis of Mesothelioma

1. Imaging

- Chest Radiography

o Unilateral Pleural effusion

o Localised mass

o Lung encasement by tumour rind

o Diffuse lobular masses

o Plaques

- Computed tomography

o Fluid only:74%

o Localised or diffuse pleural mass: 92%

o Thickening of interlobular fissure: 86%

11

o Chest wall invasion 18%

o Signs of asbestos exposure:20%

- Magnetic resonance imaging

o Can be helpful in planning of radiotherapy to localised disease

o Good for assessing tumour extent and chest wall invasion

- Positron emission tomography

o Useful for assessing tumour likelihood, and extent

o Can be helpful in staging

2. Cytopathology

o Pleural or ascetic fluid is often blood-stained

o Malignant cells seen in fluid: 33-84%

o Fine needle aspiration sampling of masses is useful

o Characteristic pattern of staining (eg, EMA positive, CEA negative)

3. Histopathology

o Closed biopsy: 30-50% positive

o Direct thoracoscopic biopsy:98%

o Immunohistochemistry (EMA, WT1, calretinin positive and CEA, CD15, Ber-EP4 negative)

4. Blood tests

o Non-specific features of malignancy (anemia, thrombocytosis, raised ESR)

o Abnormal liver function tests

o Serum mesothelin-related protein

5. Pulmonary function tests

o Restrictive pattern with increased maximum expiratory flow rates

o Volume changes vary according to amount of pleural fluid

o Changes in FVC are an accurate guide to disease progression or regression

Adapted from The Lancet, Vol. 366, Robinson BW; Musk AW; Lake RA, Malignant Mesothelioma, P 397-408, Copyright 2015,

with permission from Elsevier.

1.1.7 Current Management

Management of MPM continues to be very challenging. The limited number of patients, the

difficulty in objectively assessing responses and the lack of good quality randomised control

trials are challenges in studying effective therapies (Mossman et al. 2013).

12

Currently, the management of MPM is a multidisciplinary treatment approach in most centres,

and is based on the extent of disease, patient’s overall health condition and comorbidities and

patient’s desire for an aggressive treatment. Current guidelines recommend surgery for patients

with clinical stages I-III as part of the multimodal approach. For patients with stage IV disease,

sarcomatoid histology and medically inoperable stage I-III, chemotherapy is the treatment of

choice (Ettinger et al. 2012). Because most patients present with advanced disease, the mainstay

of therapy is chemotherapy (Raja, Murthy, and Mason 2011).

1.1.7.1 Surgery

Surgery alone is unable to improve survival and needs to be combined within a multimodality

approach. The goal of surgery is macroscopic complete resection (MCR). There currently are

two major surgical approaches that can achieve MCR: extrapleural pneumonectomy (EPP) and

pleurectomy/decortication (P/D). EPP is a well standardized surgical technique and consists of

en bloc resection of the parietal and visceral pleura with the ipsilateral lung, pericardium and

diaphragm. P/D is not standardized everywhere, in some centres P/D is defined as macroscopic

tumor removal with pleurectomy of the parietal pleura and decortication of the visceral pleura,

while others include resection of pericardium and diaphragm. This latter approach is now

nominated “extended” P/D (EPD) (Opitz 2014).

The optimal surgical approach is not clear and the role of EPP has been the subject of debate

after the publication of the MARS I trial, a multicentre randomised feasibility study. The MARS

I trial, in contrast to other phase II studies, concluded that EPP offers no benefit and possibly

harms patients compared to P/D. However, the study included only 16 patients in the EPP arm

13

and was designed as a feasibility study (Opitz 2014; Treasure et al. 2011). Recently, the initial

analysis of the IASLC reported a survival advantage in patients undergoing EPP compared to

P/D (Valerie W. Rusch et al. 2012). In conclusion, the role of EPP versus P/D remains highly

controversial and there is no clear indication as to which operation is more advantageous.

Furthermore, during the IMIG meeting in Boston in 2012, the role of surgery including P/D and

EPP in the treatment of MPM was reviewed and the attendees of the meeting agreed on the

following points:

A) Surgical MCR and control of micrometastatic disease play a vital role in the

multimodality treatment of MPM;

B) Surgical cytoreduction is indicated when MCR is deemed achievable;

C) The type of surgery (EPP or P/D) depends on clinical factors and on individual surgical

judgement and expertise;

D) All patients with MPM should initially be evaluated in a multidisciplinary setting;

E) Clinical staging should be performed before therapy;

F) The histologic subtype should be identified by tissue biopsy before initiation of therapy

(V. Rusch et al. 2013).

14

1.1.7.2 Chemotherapy

Patients that are not a good candidate for surgery are treated with chemotherapy. The

combination an antifolate drug such as pemetrexed and cisplatin achieves the best overall

survival and quality of life, and is currently the first-line chemotherapy regimen. Cisplatin is a

platinum-based drug that causes apoptosis through the cross-linking of DNA (Tanida et al.

2012). Pemetrexed, chemically similar to folic acid, works by inhibiting three enzymes involved

in purine and pyrimidines synthesis: thymidylate synthase, dihydrofolate reductase and

glycinamide ribonucleotide formyltransferase (Mita et al. 2006). The combination is the most

common regimen used for non-surgical candidates but also in the neo-adjuvant or adjuvant

setting. In a phase III study by Vogelzang et al (Vogelzang et al. 2003), the median survival for

patients who received pemetrexed and cisplatin was significantly longer than in patients

receiving only cisplatin (12.1 vs 9.3 months). The time to progression was also longer in the

combination group (5.7 vs. 3.9 months), as was the objective response rate (41 vs. 17 percent).

These differences were more striking after supplementation with folic acid and vitamin B12

during therapy and this is currently the standard of care. Second line chemotherapeutic agents

that have shown moderate increase in survival times are gemcitabine (Garland 2011; Castagneto

et al. 2005; Nowak et al. 2002) and vinorelbine (Garland 2011; Stebbing et al. 2009; Muers et al.

2008) alone or in combination with cisplatin.

1.1.7.3 Radiotherapy

Radiation has been typically used for gross tumor control as palliative intent or as part of

multimodality treatment for adjuvant local control in the postoperative setting. Adjuvant

15

radiotherapy seems to reduce local recurrence rates in some series, but its role in the

management of MPM remains unclear (Baldini 2004; V W Rusch et al. 2001).

There are some particular challenges with radiation therapy in MPM, such as the large target

volume and the presence of vital structures in the field. It is particularly challenging when the

lung is in place, as is the case when radiation follows P/D. Therefore, the main application of

radiotherapy is postoperatively following EPP since the lung is removed (Kotova, Wong, and

Cameron 2015). However, some groups have used radiation after P/D and report acceptable

toxicity profile (Minatel et al. 2014; Rosenzweig et al. 2012).

Various fractionation modalities have been used, but the most successful and most accepted are

3D conformal (3D-CRT) and intensity modulated radiotherapy (IMRT). The combination of EPP

and IMRT has been very successful at controlling local disease, but ultimately most patients will

present with metastatic disease (Ahamad et al. 2003). In the palliative setting, radiotherapy is

used for pain control and prevention or relief of obstructive symptoms (Stahel et al. 2010).

Finally, newer approaches such as the role of preoperative radiation was evaluated in the

feasibility study, Surgery for Mesothelioma After Radiation Therapy (SMART) trial. In this

study, high dose hypofractionated radiation was given to the hemi thorax 1 week prior to EPP.

Initial results showed that the protocol is feasible and reported promising survival data (Cho et

al. 2014).

16

1.1.7.3.1 SMART Trial

The “SMART” approach for resectable malignant pleural mesothelioma, was developed by Dr.

de Perrot and Dr. Cho at the University of Toronto. SMART was conceived after they observed

successful local control with hemithoracic radiation after EPP without direct effect in controlling

distant failures. The most common site of distant failure were the abdominal peritoneal cavity

and contralateral lung. They hypothesized that a mechanism of failure could be inadvertent

tumor spillage at the time of EPP. In this context, neoadjuvant radiotherapy was developed with

the presumption that its tumoricidal/tumorostatic effect on tumor cells could prevent the ability

of clonogens to proliferate in distant places if intraoperative spillage occurred. To limit the risk

of toxicity of the lung, they proposed the protocol with a short accelerated course of high-dose

hypofractionated hemithoracic radiation followed by EPP.

They conducted a seamless phase I/II feasibility study on surgically resectable MPM. The study

aim was to evaluate the feasibility of SMART. The primary endpoint was that the proportion of

patients treated with grade 5 (GS5) treatment-related mortality should not exceed 8%. Secondary

aims included morbidity, local and distant recurrence, disease free survival and overall survival

rates. Radiation dose to the target volume was 25 Gy in five daily fractions with a boost of 5Gy

to the tumor and tract sites. IMRT technique was used. All patients underwent EPP within 1

week of completion of IMRT. Cases with lymph node involvement were offered adjuvant

chemotherapy with cisplatin and an antifolate agent such as pemetrexed after EPP.

Initial results were published with 25 patients completing IMRT and EPP. The study showed that

the protocol is feasible without elevated perioperative morbidity and mortality. There was one

patient who developed grade 5 toxicity. After a median follow up of 23 months overall survival

reached 58% at 3 years. For the epithelial subtype alone the study reported the very promising

17

result of 84% survival at 3 years. While these results are encouraging, further study with a larger

number of patients is still in progress.

The authors postulated that the short course of radiation induces a tumoricidal/tumorostatic effect

that prevents implantation of tumor cells to distant sites after EPP. Furthermore, based on

growing evidence (Y. Lee et al. 2009; Levy et al. 2013; Kalbasi et al. 2013), they suggested that

hypofractionated radiation, not only has a direct cytotoxic effect, but also activates the immune

system against the tumor. Thus, the protocol of radiation followed by EPP may have an

important beneficial impact on the immune system by activating cytotoxic T cells and by

removing the immunosuppressive environment created by the tumor (Cho et al. 2014).

Figure 2. SMART Study schema. RT, radiotherapy.

Reproduced With kind permission from Wolters Kluwer Health, Inc. Journal of Thoracic Oncology, A Feasibility Study

Evaluating Surgery for Mesothelioma After Radiation Therapy: The “SMART” Approach for Resectable Malignant Pleural

Mesothelioma, volume 9, issue 3, p. 397-402, 2014, Cho, B.C., Jon Feld, Natasha Leighl, et al., figure 1.

18

1.1.8 Prospective treatment Options for MPM

Based on the understanding of the biology of mesothelioma there are many trials evaluating

newer therapeutic approaches. Many of these trials involve novel drugs affecting the molecular

signalling of tumor cells or balancing the immune system toward an antitumor profile. Some of

the targeted mechanisms include: Tyrosine kinase inhibitors, antibody conjugated toxins,

immune checkpoint inhibitors, gene therapy and tumor vaccines (Kotova, Wong, and Cameron

2015).

1.1.8.1 Tyrosine Kinase inhibitors

Several studies of MPM tumors have shown overexpression of protein targets, specifically

epidermal growth factor receptor (EGFR) (Mezzapelle et al. 2013; Okuda et al. 2008) and

vascular endothelial growth factor (VEFG) (Ohta et al. 1999; Demirag et al. 2005). EGFR a

tyrosine kinase receptor, through its activation promotes cellular proliferation and angiogenesis

and interferes with apoptosis. In-vitro inhibition of EGFR signaling in MPM cells leads to

decrease cell proliferation (Jänne et al. 2002). Although the effect was very promising in vitro,

several clinical trials evaluating drugs targeting the intracellular tyrosine kinase such as gefitinib

and erlotinib have failed to demonstrate any benefit in survival (Govindan et al. 2005; Garland et

al. 2007). However, in non-small cell lung cancer patients, treatment with chemotherapy and anti

EGFR monoclonal antibodies (cetuximab), targeting the extracellular domain of EGFR,

significantly improved overall survival compared to chemotherapy alone (Pirker et al. 2009). The

phase II Mesomab trial (NCT00551252 ) is evaluating cetuximab in combination with

chemotherapy in MPM patients.

19

VEGF is a powerful endothelial cell-specific mitogen associated with tumor neovascularisation

and cell proliferation. Expression of VEGF and its receptor VEGFR-1, -2 and -3 have been

demonstrated in human mesothelioma cells lines (König et al. 2000; Masood et al. 2003). VEGF

overexpression in MPM tumor samples is associated with poor prognosis (Demirag et al. 2005).

In-vitro studies showed that inhibition of VEGF greatly reduces MPM cell viability (Masood et

al. 2003). Hence, inhibition of this pathway has received great attention as a potential anti-

neoplastic therapy. Results of numerous clinical trials evaluating VEGF inhibition in MPM have

failed to show any significant difference in survival (Jahan et al. 2012; Dowell et al. 2012; Papa

et al. 2013; Ceresoli et al. 2013). However, an ongoing clinical trial (NCT00651456) presented at

the 2015 American Society of Clinical Oncology Annual Meeting presented promising results in

patients treated with bevacizumab. The group showed significant longer survival in patients with

MPM treated with Pemetrexed, Cisplatin and Bevacizumab compared to patients treated with

Pemetrexed and Cisplatin only (Zalcman et al. 2015).

1.1.8.2 Antibody conjugated toxins

Mesothelin is a protein present on normal mesothelium and overexpressed on epithelial cancer

like mesothelioma, ovarian cancer, lung adenocarcinoma and pancreatic adenocarcinoma (Chang

et al. 1992). Its restricted expression on normal tissues and overexpression on neoplastic cells

make mesothelin a good target for antibody based therapy. Amatuximab (MORAb-009) is a

chimeric high-affinity monoclonal IgG1/k antibody targeting mesothelin. In-vitro, after binding

mesothelin the antibody is internalized and elicits antibody-dependent cellular cytotoxicity

(ADCC). Preclinical studies with xenografts, combination treatment with amatuximab and

chemotherapy was more effective than either amatuximab or chemotherapy alone (Hassan et al.

20

2007). A phase I study demonstrated amatuximab is well tolerated (Hassan et al. 2010). In a

phase II clinical trial, amatuximab was combined with pemetrexed and cisplatin in patients with

unresectable MPM. The study did not meet the primary endpoint of 3-month improvement in

progression free survival over historical controls. However, the authors highlighted the median

OS of 14.8 months in this particular population with 87% of patients with stage III/IV disease.

Together with the fact that a third of the patients were alive at the time of analysis, the authors

suggested that Amatuximab potentially had an antitumor activity (Hassan et al. 2014).

CRS-207 is a live-attenuated double deleted Listeria monocytogenes strain that was engineered

to express human mesothelin. CRS-207 induced mesothelin-specific T-cell responses that

correlated with regression of murine tumors in preclinical studies. A phase I clinical trial

determined that it was well tolerated and demonstrated an induction of tumor antigen-specific T

cells responses in patients with advanced cancer. A CD8 T cell specific for mesothelin response

was induced in six out of 10 patients but did not correlate with clinical response (Le et al. 2012).

SS1P is an immunotoxin consisting of the variable fragment of an anti-mesothelin antibody

linked to a truncated form of Pseudomonas exotoxin A (PE38). When SS1P binds its target,

PE38 is internalized and kill cells by ADP ribosylation and inactivation of elongation factor 2

(EF-2) and apoptosis (Pastan et al. 2007). A phase I trial showed that SS1P was well tolerated

and had modest clinical activity. However, many patients developed neutralizing antibodies that

prevented additional therapy (Kreitman et al. 2009). A second study showed better results after

combining SS1P with pentostatin and cyclophosphamide, to induce an immunosuppressive state

and abrogate the production of neutralizing antibodies (Hassan et al. 2013).

Interleukin-4 (IL-4) receptors are present on several human tumors including mesothelioma,

making it a potential clinical target. A recombinant IL4 toxin called cpIL-4-PE was designed

21

with fragments of IL-4 fused with a variant form of the exotoxin PE38 (Puri et al. 1996). This

IL-4 toxin binds mesothelioma cells and inhibit protein synthesis in vitro. Pre-clinical studies in

mice are promising and show reduced tumor volumes and prolonged survival in mice treated

with the toxin compared to controls (Beseth et al. 2004).

1.1.8.3 Mesothelin-specific Chimeric Antigen Receptor T cells

Tumor antigen specific T cells have been engineered by the introduction of chimeric antigen

receptors (CARs) that have antibody-based external receptor structures and cytosolic domains

that encode signal transduction modules of the T cell receptor. This concept was first developed

by Dr. Eshhar and his group created the first functional CAR T cells by 1989 (Gross et al. 1989).

The recognition element is derived from an antibody variable region and this retargets T cells in

an MHC unrestricted manner and are not patient specific, thus the same chimeric antigen

receptor can be used for multiple patients. Clinical trials have documented safety but poor in

vivo persistence and expression of the transgene (Kershaw et al. 2006; Till et al. 2008). Second

and third generation CAR T cells have been engineered to overcome these shortcomings and

incorporate costimulatory signaling domains into the signaling module (Till et al. 2012; Heiblig

et al. 2015).

The application of CAR T cells to treat solid malignancies has been limited in part due to the

potential of CAR-based therapies to cause on-target off-tumor toxicity through recognition of

normal cells that express the target antigens (Lamers et al. 2013). To circumvent this toxicity

several groups have incorporated safety genes such as inducible caspase 9 transgene with

variable results (Di Stasi et al. 2011).

22

Mesothelin is overexpressed in the majority of MPM but it is also expressed at low level on

normal peritoneal, pleural and pericardial mesothelial surfaces. In a preclinical model Carpenito

et al., (Carpenito et al. 2009; Zhao et al. 2010) evaluated mRNA electroporated mesothelin CAR

T cells (CARTmeso cells) and observed potent antitumor effects against established tumor

xenografts. mRNA electroporated T cells transiently express the mesothelin-targeting CAR, thus

reducing off-tumor toxicity. The same group reported the use of CARTmeso cells is feasible and

safe in humans and reported two cases from an ongoing trial (Gregory L Beatty et al. 2014).

Clinical and laboratory evidence of antitumor activity was demonstrated in both patients. Their

data support the feasibility of CAR T cells as a novel strategy for the treatment of patients with

solid malignancies such as MPM.

1.1.8.4 Gene Therapy

1.1.8.4.1 Suicide Gene Therapy

In this approach, tumor cells are transduced with genes that encode for proteins that metabolize

prodrugs into toxic metabolites. These metabolites accumulate in tumor cells and leads to its

death or “suicide”. A common gene is thymidine kinase gene (HSVtk) from the herpes simplex

virus-1. Thymidine kinase metabolizes the nontoxic antiviral ganciclovir into GCV-

monophosphate which is then catalyzed by cell kinases into ganciclovir triphosphate. This

metabolite is a competitive inhibitor of DNA polymerase and disrupts DNA synthesis (Markham

and Faulds 1994; Vachani et al. 2010). A phase 1 clinical trial of adenovirus gene therapy

(Ad.HSVtk/GCV) in patients with MPM was done to evaluate safety, immunologic responses,

transgene expression and clinical responses. A single intrapleural dose of the vector was given

23

followed by GCV I.V. twice daily for 14 days. The vector was well tolerated and deemed safe.

23/30 patients had demonstrated gene transfer. Post treatment antibody responses against the

tumors were seen and proliferative T cell responses were generated in serum and pleural fluid.

Clinical responses were seen and 2 patients showed long term survival (7 and 10 years) (Sterman

et al. 2005). In another phase1 clinical trial, ovarian carcinoma cells transfected with the HSVtk

gene (PA1-STK) were infused intrapleurally followed by GCV. Some of the cells were followed

with 99Tc and it was shown that the labeled cells adhered preferentially to intrapleural

mesothelioma deposits. The transfected cells were thought to have exerted a bystander effect on

mesothelial cells–the killing of neighboring cells not transduced with the vector. Patients showed

minimal side effects and the authors concluded this therapy is feasible in humans (Harrison et al.

2000).

1.1.8.4.2 Cytokine Gene Therapy

This therapy is based on the administration of viral vectors encoding cytokine genes that may

lead to high expression of immunostimulatory cytokines. Some of the cytokines may have a

cytotoxic effect or activate the immune system. There are several trials that evaluated

administration of IL-2 with good results in phase I and II trials (Astoul et al. 1998; Tan et al.).

Recently gene therapy has centered on IFN which plays a central role in the activation of the

immune system and may have a direct anti-tumor cytotoxic effect (Sterman et al. 2011; Odaka et

al. 2001; Sterman et al. 2007).

24

1.1.8.5 Tumor Vaccines

Dendritic cell vaccines are used to improve effective tumor antigen presentation and successfully

activate the immune system. Dendritic cells are the most potent antigen presenting cells.

Preclinical data has shown encouraging results and may be a valuable strategy in cancers like

mesothelioma (Palucka and Banchereau 2012; Hegmans et al. 2010). Besides autologous tumor

lysates, calretinin, mesothelin and WT-1 have been used as antigens. In the case of WT-1,

measurable CD4 and CD8 T cells responses were elicited, but there were no clinical response

(Krug et al. 2010). With autologous tumor antigens in a phase I clinical trial, the vaccine was

well tolerated and there were several partial responses (Hegmans et al. 2010). Currently, there

are several phase I-II trials running using WT-1, Mesothelin and 5T4 as vaccine antigens

(Kotova, Wong, and Cameron 2015).

1.1.8.6 Immune Checkpoint Inhibitors

Antibodies that modulates the immune system by binding to checkpoint molecules and shifting

the immunes system towards an anti-cancer response are starting to be used in MPM. Immune

checkpoints are pathways that regulate and modulate immune responses and are crucial for

maintaining self-tolerance. Some of these checkpoints are triggered by ligand-receptor

interactions leading to the potential for a therapeutic target, by blocking these interactions

(Brahmer and Pardoll 2013).

Cytotoxic T lymphocyte antigen-4 (CTLA-4), also known as CD152 is one of the receptors most

actively studied in the context of cancer immunotherapy (Postow, Harding, and Wolchok 2012).

25

Briefly, CTLA-4 receptor raises the threshold for T lymphocyte activation by sequestering the

co-stimulatory signals provided by CD80 and CD86 present on antigen-presenting cells. By

blocking the CTLA-4 receptor with antibodies, the negative regulatory effect of the receptor can

be reversed and a therapeutic response against tumor cells can be induced. We have shown in our

lab in a mouse model of mesothelioma that blockade of CTLA-4 demonstrates an anticancer

effect and is correlated with inhibition of cancer cell repopulation when combined with

chemotherapy (L. Wu et al. 2012). A phase II clinical trial evaluated tremelimumab, an anti-

CTLA-4 antibody. The study enrolled chemotherapy-resistant advanced mesothelioma patients

and showed clinical responses in 2/29 patients, stabilization in 9/29 and overall survival at one

year was 48% (Calabrò et al. 2013). Currently there are two fully human antibodies, ipilimumab

and tremelimumab, both block CTLA-4 interaction with B7 ligands. The antibodies have

undergone extensive study in melanoma (Hodi et al. 2010; Sanford 2012; Ascierto 2013; Larkin

et al. 2015) and ipilimumab was approved to treat patients with advanced melanoma in the US,

Canada and the European Union. Tremelimumab was approved recently by the FDA to treat

MPM.

The other immune checkpoint studied for cancer immunotherapy is the programmed death 1

(PD-1) pathway. The pathway PD1 and programmed death ligand 1 (PD-L1) limits the activity

of T cells in peripheral tissues during inflammation. PD-1 is found on the surface of T cells and

PD-L1 is expressed on the surface of tumor cells (Zou and Chen 2008). Expression of PD-L1

was first demonstrated in murine mesothelioma (Currie et al. 2009) and later on human

mesothelioma specimens. Expression of PD-L1 was seen mostly on sarcomatoid and biphasic

subtype of MPM and was associated with poor prognosis in 2 recent publications (Mansfield et

al. 2014; Cedrés et al. 2015). Studies in our lab demonstrated no effect of anti PD-1 mAb alone

on tumor growth in a subcutaneous murine mesothelioma model. However, we have observed

26

dramatic tumor shrinkage when combined with local radiotherapy (unpublished data). Clinical

trials in melanoma and lung cancer have shown durable clinical activity with anti PD-1

immunotherapy (Topalian et al. 2012; Lipson et al. 2013; Hamid et al. 2013). Currently, there are

no clinical trials evaluating the role of anti PD-1 in MPM, but it remains a promising option.

1.2 Tumor Immunity

1.2.1 The immune response

The immune system consists of two integrated systems, the innate and the adaptive system. They

each consist of both cellular and non-cellular components. The innate system provides us with a

rapid immune response against great variety of pathogens by recognizing evolutionary conserved

molecules (O. Krysko et al. 2013). Adaptive immunity involves an orchestrated response

involving cellular and humoral components. The adaptive immune system requires more time

but is more specific. Furthermore, the adaptive immune system generates an antigen specific

response against pathogen molecules not previously encountered by the host and in many cases

results in immunological memory (Alberts et al. 2002). This project is focused on the adaptive

system as it provides a specific response as well as immune memory. In cancer a successful

adaptive response can specifically target cancerous cells and prevent the recurrence of disease

(D. S. Chen and Mellman 2013).

27

1.2.2 Adaptive immunity

Adaptive immunity is predominantly comprised of B and T cell lymphocytes. There are two

main branches of the adaptive immune response: the humoral immune response mediated by B

cells, and the cell-mediated immune response directed by T cells. The humoral immune response

is characterized by the production of antibodies. Antibodies bind to their targets and mediate the

pathogen clearance or inactivation. Cellular mediated responses include direct killing of target

cells such as virally infected or cancerous cells by T cells or NK cells, as wells as activation of

other cells through production of cytokines by T cells (Norvell 2013).

1.2.2.1 T cells

T cells, derive their name from their maturation in the thymus and are broadly classified into

cytotoxic, helper and regulatory T cells. In cancer, T cells are the principal mediators of

antitumor immunity. CD8+ T cells recognize pathogen peptides loaded on the major

histocompatibility complex (MHC) class I molecules by antigen presenting cells. Naïve CD8 T

cells can differentiate into cytotoxic T cells (CTL) that recognize and kill virally infected or

cancerous cells. CTLs kill their targets through perforin mediated cytotoxicity and Fas/FasL

interaction (Nagata and Golstein 1995; Henkart 1994; Seki et al. 2002). There are numerous

studies that have examined and found a positive correlation between the presence and number of

CD8+ tumor-infiltrating T cells (TILs) and good prognosis. This positive association has been

reported in colorectal cancer, ovarian and breast cancer among others (Bachmayr-Heyda et al.

2013; Matkowski et al. 2009; Nosho et al. 2010). Our group, reported that high levels of CD8+

TILs between cycles of chemotherapy is associated with longer progression-free survival as well

28

as delayed recurrence in MPM in a murine model (Licun Wu et al. 2011). Furthermore, our

group reported the association in MPM patients of high levels of CD8+ tumor infiltrating

lymphocytes with better survival in patients undergoing EPP (Anraku et al. 2008). The

importance of CD8 T cells in immune therapy is highlighted by the attempts to activate and

increase their number in order to improve survival in patients (Dudley et al. 2002; Fourcade et al.

2010).

1.2.3 The T cell immune response

For an anticancer immune response to effectively kill cancer cells, a series of events must occur

to ultimately activate and expand T cells.

In the first step immature dendritic cells (DC) uptake tumor antigens present in the extracellular

environment primarily through pinocytosis. The up-taken antigens are then cleaved and

processed for loading onto the MHC molecules. If DCs receive specific signals or sense

“danger” they upregulate costimulatory molecules and the chemokine receptor CCR7. CCR7

recognizes the chemokines produce by lymphoid tissue CCL19 and CCL21 and leads the DC to

the T-cell zones of the local lymph nodes. The role of DC in the lymph nodes is to present

antigens to specific T lymphocytes. The signals involved in DC maturation may include

pathogen-associated molecular patterns (PAMPs) or damage-associated molecular pattern

molecules (DAMPs) and proinflammatory cytokines and factors released by dying tumor cells.

In the second step, DCs present the antigens on MHC I and MHC II molecules to naïve T cells

resulting in the priming and activation of effector T cell responses against tumor specific

antigens. To activate T cells three stimulatory events must occur. The first signal involves the

29

specific peptide-MHC complex and T cell receptor (TCR) interaction which is stabilized by

either CD4 or CD8 molecules on T cells. The second signal involves the costimulatory

interaction of B7.1 and B7.2 on DCs binding to CD28 on T cells. Additional costimulatory

interactions include OX40L-OX40, CD70-CD27, CD40-CD40L, CD137L-CD137, ICOSL-ICOS

(Driessens, Kline, and Gajewski 2009). The last signal involves cytokines secreted by APCs that

differentiate T cells into one of many T cell subsets that include but are not limited to CD8

cytotoxic, Th1, Th2, Th17, and T regulatory cells. The encounter with an specific antigen in the

presence of a co-stimulatory signal triggers T cell proliferation and, induces the synthesis of IL-2

and the α chain of the IL-2 receptor (CD25). Association of the α chain with the β and γ chains

forms a high affinity IL-2 receptor, allowing T cells to respond to very low concentrations of IL-

2. IL-2 then stimulates T cell proliferation, differentiation and survival. After 4-5 days of rapid

proliferation induced by IL-2, activated T cells differentiate into effector T cells that can

synthesize all the molecules required for their cytotoxic functions.

In the next step the activated effector T cells traffics to the tumor where it recognizes and binds

to cancer cells through interaction of the TCR and the antigen MHC complex. This interaction

results in immune attack without the need for co-stimulation. T cells will then kill the target

cancer cells. Killing of the cancer cell releases additional tumor associated antigens and this in

turn can increase the intensity of the response against the tumor. The antitumor immune response

will be defined at this point, with a balance between the ratio of effector T cells versus regulatory

T cells. Experimental evidence has shown that tumor rejection requires T cells that have

functionally differentiated to become CD4+ Th1 and CD8+ cytotoxic T cells (CTL) (Nishimura

et al. 2000).

30

1.2.4 Tumor Escape

The idea of immune surveillance for eradicating nascent transformed cells before they are

clinically detected was first proposed by Ehrlich in the early 20th century (Ehrlich 1909). Fifty

years later, experimental evidence that tumors could be repressed by the immune system came

from tumor transplantation models. This led to the formal hypothesis of “cancer

immunosurveillance” by Burnet and Thomas (Burnet 1957). Both speculated that lymphocytes

acted as sentinels in recognizing and eliminating continuously arising, nascent transformed cells

(Burnet 1970). However, subsequent experiments based on experimental immunosuppression

(Kaplan 1971; Stutman 1975) or using nude mice (O Stutman 1974; Osias Stutman 1979) failed

to prove the immunosurveillance hypothesis at that time and led to its abandonment. It was until

the 1990s when experimental animal models using knockout mice validated the existence of

cancer immune surveillance in both chemically induced and spontaneous tumors. At the same

time the central roles of T cells, NK, NKT, Interferons and perforin were clarified in cancer

immune surveillance (Dighe et al. 1994; Russell and Ley 2002; van den Broek et al. 1996;

Shankaran et al. 2001).

Finally, the current concept of cancer immunoediting leading from immune surveillance to

immune escape was proposed. Three essential phases were proposed by the Schreiber group

(Dunn et al. 2002): elimination; equilibrium; and escape. Briefly, in the elimination phase,

transformed cells can be eliminated by immune effector cells such as NK and T cells. Tumor

cells are recognized initially by NK, NKT or T cells which are then stimulated to produce IFN γ

which will lead to accumulation and activation of immune cells and eventually tumor cell lysis.

In the equilibrium phase, the host immune system and tumor cells that have survived the

elimination process enter into a dynamic equilibrium. In this phase, immune cells exert potent

31

selection pressure on the tumor cells that is enough to contain, but not fully eliminate. During

this period of selection new variants with different mutations arise and provide them with

increased resistance to immune attack. Lastly, in the escape process, tumor cells that have

acquired resistance to immunologic detection or elimination begin to expand in an uncontrolled

manner that may eventually lead to clinical disease. Eventually, during tumor progression,

tumor-derived soluble factors can induce several mechanism for escape from immune attack in

the tumor microenvironment (R. Kim, Emi, and Tanabe 2007).

As described previously, there is significant complexity for mounting an antitumor immune

response, among other factors, the priming and effector phases are separated by time and space.

Priming occurs in lymph nodes, the effector functions must operate within the tumor mass. There

are several obstacles that the system must overcome. During priming, the lack of “danger”

signals from innate immune cells, poor recruitment of DCs for cross-presentation, and

inadequate expression of costimulatory ligands on tumor cells or APCs can hinder the immune

response. Furthermore, during the effector phase there may be inadequate recruitment of

lymphocytes due to abnormal blood vessels and cytokines, activation of inhibitory receptors on T

cells such as CTLA-4 and PD-1, extrinsic suppressive cells (TREGs, myeloid-derived

suppressive cells-MDSC), metabolic inhibitors (IDO, arginase) and inhibitory cytokines (IL-10,

TGF-β) (Gajewski et al. 2011). All of the previous can limit the impact of the T cell response on

the tumor and eventually lead to progression of clinical disease and if left unchecked may result

in the death of the host.

32

1.3 Radiation Therapy of tumors and the Immune System

1.3.1 Brief history

In November of 1895, Wilhelm Conrad Rӧntgen described the x-ray and very soon after,

potential applications were recognized in different fields. Shortly after in January 1896, Emil

Grubbé treated a woman who suffered of an open inoperable carcinoma of the breast. The same

year, Despeignes was the first to publish his results using x-rays to treat gastric carcinoma

(Buschke 1958). Radiation therapy increased in popularity and advances have continued to the

present.

1.3.2 Radiation and its interaction with matter

When radiation interacts with matter, there is an absorption of energy from radiation and this

may lead to excitation or ionization. Excitation occurs when an electron in an atom is moved to a

higher energy level. Ionization occurs when there is enough energy to eject the electron from the

atom. In the latter case, radiation is called ionizing. Gamma and x-rays are example of ionization

radiation. Radiation can be classified as directly or indirectly ionizing. Charged particles, such as

electrons or protons, are examples of directly ionizing. Provided they have enough energy they

can disrupt the atoms of the structure they pass through and produce chemical and biologic

changes. On the other hand x-rays are indirectly ionizing; they do not disrupt the structure of the

atoms by themselves but they are absorbed in the material and give up their energy to produce

fast moving charged particles (electrons) that can in turn produce damage (Hall and Giaccia

2006).

33

Gamma and x-rays do not differ in nature or in properties, and can be considered from two

different standpoints, as electromagnetic waves or as a stream of photons or packets of energy.

Like radio waves and visible light, gamma and X-rays are forms of electromagnetic radiation. X-

rays occupy the short wavelength end of the electromagnetic spectrum. Also, x-rays can be

considered as packets of energy called photons. When x-ray photons are absorbed by matter at

high energies as when used in radiotherapy the Compton Process will dominate. In this process

the photon will interact with a “free” electron and part of the energy of the photon is given to the

electron as kinetic energy. The photon will continue deflected from its original path and with less

energy. The result is a photon of reduced energy and a fast electron. Electrons can then ionize

other atoms, break chemical bonds and initiate the change of events that will result in biological

damage (Hall and Giaccia 2006).

The main biologic effects of radiation results from damage to DNA. Radiation interacts with

atoms in the cells, mainly water and produces free radicals (hydroxyl radical - OH▪) that can

diffuse and reach DNA. It is estimated that 2/3 of the x-ray damage to DNA in cells is caused by

the hydroxyl radical. Estimations suggest that 1-2 Gy of x-ray radiation can result in 105

ionization events per cells, resulting in 1000-2000 single-stranded DNA breaks (SSB) and 40

double-stranded breaks (DSB) (Lewanski and Gullick 2001). SSB are of little biologic

consequence, as they are repaired readily by the cell mechanisms. However, DSBs are the most

important lesion and may result in cell killing (Radford 1985).

34

1.3.3 Factors affecting the cellular response to radiation

The final outcome in a cell after being exposed to radiation will depend mainly on the stage of

the cell cycle but also on the presence of free radical scavengers and biomolecules (oxygen).

Cells are most sensitive at or just before mitosis. Resistance is greatest in the latter part of the S

phase, as DNA damage can be repaired more rapidly at this stage (Pateras et al. 2015). Cells that

divide frequently like tumor cells or lymphocytes are more radiosensitive than those that divide

rarely such as nerve and muscle cells. In the case of a tumor, where cell division is not

synchronized, not all cells will be successfully eliminated.

The presence or absence of oxygen will influence the biologic effect of x-rays. Oxygen at the

time of radiation fixes ionization damage and makes it more difficult to repair. If cells are in

hypoxic condition they will become more resistant to radiation (Palcic and Skarsgard 1984). A

clinical study used hyperbaric chamber trying to improve the effect of radiation. They reported a

slightly improved tumor control compared to radiotherapy on normoxic conditions, but the

treatment proved to be costly and difficult to administer (Overgaard and Horsman 1996).

1.3.4 Cellular Sensing and Response to Radiation

Ionizing radiation damage have effects on transcription, DNA synthesis, cell cycle regulation and

may trigger apoptosis or cell death (Valerie and Povirk 2003). Radiation induced DNA damage,

such as DSB, is a lethal DNA lesion. However, cells are equipped with sensors that recognize

these lesions immediately after formation and a signaling cascade is started that ultimately may

result in cell cycle arrest, giving the cell time for repair if the damage is not extensive, either by

35

homologous recombination or non-homologous end-joining pathways (Valerie and Povirk 2003).

The most important sensors are ataxia-telangiectaxia mutated (ATM) and the MRN complex

(Mre11, Rad50 and Nbs1) (Lavin 2007; J.-H. Lee and Paull 2004). ATM is activated through a

functional MRN complex, which recognizes and migrates to DSBs after ionizing radiation

(Lavin 2007). The most important proteins activated by ATM are surveillance proteins, such as

p53 (Canman et al. 1998), CHK2 (Matsuoka et al. 2000) and DNA-protein kinase (B. P. C. Chen

et al. 2007). CHK2 phosphorylates in turn p53 (Shieh et al. 2000) leading to a release of p53

from MDM2. The dissociation of MDM2-p53, results in stabilization of p53. ATM also directly

phosphorylate p53 increasing its activity (Canman et al. 1998). As a results p53 translocate into

the nucleus and binds target genes, such as p21. The upregulation of p21 ultimately results in cell

cycle arrest at the G1/S checkpoint through inhibition of the Cdk2-cyclin E-PCNA complex

(Kaina, Roos, and Christmann 2010). Additionally, CHK1 via phosphorylation of Cdc25a and

Cdc25c leads to dephosphorylation of CDK2 and Cdk1-CyclinB, inducing G1/S and G2/M arrest

(Sanchez et al. 1997; Peng et al. 1997; Mailand et al. 2000).

The main apoptosis pathways activated by DNA damage involve the Fas/Caspase 8 signaling

and the apoptosome formation, both can be dependent or independent of p53/p73 (Pietsch et al.

2008). In all cases, signals converge on Caspase 3 to induce apoptosis. The upregulation of

p53/p73 in turn upregulates the fas receptor/caspase-8 apoptotic pathway (Bennett et al. 1998).

In 50% or more of all human cancers p53 is mutated (Soussi and Lozano 2005). In cells where

p53 is inactive, DNA damage activates the endogenous or mitochondrial apoptotic pathway. In

this pathway the decline of Bcl-2 leads to leakiness of the mitochondria, cytochrome c release

and activation of the apoptosome (Apaf-1, ATP, procaspase-9, and cytochrome c). Caspase 9

then cleaves caspase-3 and other downstream caspases leading to activation of caspase-activated

36

DNase (CAD). CAD ultimately cleaves DNA into the typical nucleosomal fragments (Haupt et

al. 2003).

Other factors modulate the effect of DNA damage, such as, p53 (Batista et al. 2007), Jun

kinase/p38 kinase (Hamdi et al. 2005), caspase-2(Robertson et al. 2004), NF-κB (Karin and Ben-

Neriah 2000) and Akt (Gottlieb et al. 2002). These various pathways will converge and compete.

Some transduce cell death signals, with others transducing survival signals. The summation of all

these signals are carefully regulated by the cell context and the level of DNA damage, with the

net effect of either survival, death by apoptosis or necrotic cell death in the case of high non

tolerable levels of DNA lesions (Kaina, Roos, and Christmann 2010).

1.3.5 Cell death response

Apoptosis is one type of programed cell death and is the prevalent form of cell death under

normal conditions and daily tissue regeneration. Cells undergoing apoptosis are rapidly cleared

by macrophages and dendritic cells and induce immune tolerance (Voll et al. 1997; Albert et al.

1998). Apoptosis is the main response to radiotherapy in the hematopoietic system and the

intrinsic death pathway is the major signaling mechanism (Heylmann et al. 2014; Eriksson and

Stigbrand 2010). Significantly less apoptosis is observed in cells of epithelial origin and in this

kind of cells, radiotherapy can trigger necroptosis or necrosis(Mantel et al. 2010; Vandenabeele

et al. 2010). Necroptosis is characterized by the production of ROS, lipid peroxidation, swelling

of organelles, rupture of the plasma membrane, and release of intracellular contents

(Vandenabeele et al. 2010). Also, secondary necrosis can occur when apoptotic dying cells failed

to be phagocytosed in time. This can occur when a large number of cells undergo apoptosis and

37

overwhelms the phagocytic cells, as in the context of tumor radiotherapy (Silva 2010). Mitotic

catastrophe occurs as a result of improper entry into mitosis resulting in aberrant cell division. It

is one of the main mechanism of cell death secondary to radiation induced DNA damage in cells

with defective cell cycle checkpoints and impaired DNA repair mechanisms (p53 mutations)

(Eriksson and Stigbrand 2010). Cells that entered into mitotic catastrophe may survive for days,

transit into senescence or die by delayed apoptosis, or necroptosis (Lauber et al. 2012). Mitotic

catastrophe is a delayed type of cell death, days after treatment initiation, and may explain the

slow clinical regression of solid tumors. Senescence is a condition of permanent cell cycle arrest.

Senescence secondary to radiation is observed in cells where cell cycle checkpoints are still

intact (Eriksson and Stigbrand 2010). Cell death by necrosis or necroptosis is not

immunologically silent and can trigger a potent inflammatory immune response. Necrosis

releases cellular debris and pro-inflammatory molecules into the extracellular space including

high-mobility group box 1 proteins (HMGB-1), heat-shock proteins (HSP), calreticulin and ATP

(Rock and Kono 2008; Kono and Rock 2008). These mediators can orchestrate the recruitment

of immune cells, according to the danger hypothesis proposed by Matzinger (Matzinger 1998),

and eventually this lead to activation of innate and adaptive immune system (Tesniere et al.

2008; Jonathan, Bernhard, and McKenna 1999; Illidge 1998). Therefore, the type of death,

timing of clearance, and factors released during cell death will elicit a response from the host that

will shape the state of the immune system after irradiation.

1.3.6 Immunogenic Cell Death

Cell death and necrosis in a radiated tumor may alter the activation state of the immune system.

The danger hypothesis states that dying cells release signals that aid the immune system to

38

recognize forms of non-physiological cell death. This form of cell death releasing specific

signals that stimulate adaptive immune responses has been named “immunogenic cell death”

(ICD) (Tesniere et al. 2008; Obeid et al. 2007; Golden et al. 2012; Golden and Apetoh 2015;

Kepp et al. 2011). In necrotic cell death, these signals involve HSP, HMGB-1, Calreticulin and

ATP among others. These endogenous molecules that deliver danger signals in response to stress

and trigger ICD are known as damage associated molecular patterns (DAMPs). Although the

details of ICD continue to be elucidated and other molecules cannot be excluded, 3 necessary

components have been characterized, the exposure of the endoplasmic reticulum (ER) chaperon

calreticulin (CRT) on the outer surface of the plasma membrane, the release of the non-histone

chromatin-binding HMGB1 and ATP release (Ma et al. 2010; Golden and Apetoh 2015; Obeid et

al. 2007). Calreticulin is an “eat me” signal and enables phagocytes and dendritic cells to

efficiently engulf dead cells. Calreticulin is recognized by CD91 (LRP1) positive cells

(macrophages and DCs) unless they simultaneously express the “do-not-eat me” signal CD47

(Garg et al. 2012; D. V Krysko et al. 2012; Kroemer et al. 2013). ATP is a “find me signal” and

promotes recruitment of APC by binding to the receptors, purinergic receptor P2Y, G-protein

coupled, 2 (P2Y2) and purinergic receptor P2X ligand-gated ion channel 7 (P2X7) (D. V Krysko

et al. 2012; Bezu et al. 2015). Stimulation of P2Y2 is required for monocyte attraction and

activation of the P2X7 receptors on dendritic cells activates the NALP3 inflammasome, a

multimeric danger-sensing platform that promotes and drives the secretion of IL-1β (Garg et al.

2012). This cytokine is required for the polarization of IFN-γ producing CD8+ T cells

(Ghiringhelli et al. 2009). HMGB-1 exerts its immunostimulatory effects through TLR4 and

advanced glycosylation end product-specific receptor (RAGE). Binding of HMGB-1 to these

receptors on innate immune cells such as neutrophils, macrophages and monocytes stimulates the

production of pro-inflammatory cytokines (TNF-α, IL-1, IL-6 and IL-8) (Rovere-Querini et al.

39

2004; G. Chen et al. 2004). In summary, danger signals released from necrotic cells as a result of

radiation play a role in the activation of immune responses that can tip the balance against the

tumor.

1.3.7 Tumor microenvironment

1.3.7.1 Cytokine expression

Radiotherapy has a significant effect on the modulation of immune responses and this effect is

due in part to the balance between pro-inflammatory and anti-inflammatory cytokines and

chemokines. Radiation leads to an increase in IFN-γ production (A. A. Lugade et al. 2008). IFN-

γ has pleiotropic effects in the tumor microenvironment, including the upregulation of MHC

class I and II expression (Weber and Rosenberg 1988; Dighe et al. 1994), activation of

macrophages (Boehm et al. 1997; Xie, Whisnant, and Nathan 1993), inhibition of the production

of immunosuppressive molecules (Hirte and Clark 1991), and enhancement of the secretion of

antiangiogenic chemokines (Sgadari, Angiolillo, and Tosato 1996; Arenberg et al. 1996). IFN-γ

production by infiltrating T cells enhances expression of VCAM-1 and ICAM-1 to further

enhance T cell infiltration (A. A. Lugade et al. 2008; Caldenhoven et al. 1994). Furthermore,

IFN-γ activates STAT1 which drives the expression of multiple chemokines including CXCL9

(MIG), CXCL10 (IP-10). These chemokines are potent signals for T cell activation through the

receptor CXCR3 (Burnette and Weichselbaum 2013). However, IFN-γ has also the potential to

down regulate antigen presentation by tumor cells (G L Beatty and Paterson 2000), either

through down regulation of tumor antigen protein expression of by less efficient processing of

tumor antigens (S. Morel et al. 2000). TNF-α is another cytokine upregulated after radiation (van

40

Valen et al. 1997; Rübe et al. 2004; Fedorocko, Egyed, and Vacek 2002) and mediates its effects

in concert with IL-6 and IL-1 (Desai et al. 2013). TNF- α is a cytokine with dual effects, at low

concentrations promotes tumor angiogenesis, tumor cell survival and metastasis, but at high

levels prevents tumor growth and induce the expression of adhesion molecules and increase

vascular permeability, together with IFN-γ (S. Kim et al. 2009; Lumniczky and Sáfrány 2015).

The main negative regulator of inflammations in a radiated tumor include TGF-β (Roedel et al.

2002) and IL-10. TGF-β is upregulated and activated from its latent form after radiation

(Barcellos-Hoff et al. 1994). TGF-β is another pleiotropic cytokine and play a role in

extracellular remodeling accompanied by cancer cell migration and invasion (De Wever and

Mareel 2003). TGF-β secreted locally in the tumor microenvironment after radiation modulates

the local inflammatory response, suppresses DCs and CTL functions and promotes the

infiltration of CD4 Regulatory T cells (Barcellos-Hoff et al. 1994). IL-10 another anti-

inflammatory cytokine, secreted by cancer cells and apoptotic lymphocytes and monocytes also

influence the outcome of the immune response (Gao et al. 1998; Lumniczky and Sáfrány 2015).

The understanding of the intricate signalling and effects of cytokines in the tumor

microenvironment after radiation therapy will be essential for instituting better interventions

such as immunotherapeutic interventions.

1.3.7.2 Changes in the tumor microenvironment

Tumor cells undergo a process of immune-editing, and develop several mechanisms by which

neoplastic cells can escape immune recognition and elimination. As a result the tumor

microenvironment is an immunosuppressive environment. Radiation causes important changes in

the tumour cells that have implications for its interaction with the immune system.

41

One escape mechanism of tumor cells is the reduction of MHC I molecules on the surface

leading to inadequate antigen presentation (Rabinovich, Gabrilovich, and Sotomayor 2007).

However, after irradiation MHC Class I are up-regulated in a variety of tumors, both in vitro and

in vivo (Chiriva-Internati et al.; Ciernik et al. 1999; Reits et al. 2006). Furthermore, the cellular

damage induced by radiation increases the peptide concentration and repertoire displayed in

radiated cells (Reits et al. 2006). It has been shown by Reits et al. (Reits et al. 2006) that tumor

cells with upregulated MHCI molecules are eliminated more efficiently by tumor-specific

cytotoxic T lymphocytes (CTLs). Additionally, upregulation of the death receptor CD95, in a

p53 dependent way, was reported on multiples tumor models (Sheard, Uldrijan, and Vojtesek

2003; I.-C. Park et al.). CD95 signaling is also enhanced and leads to an increase in tumor

immunogenicity, since it can improve the cytotoxic effect of CD95 ligand-expressing CD8+ T

lymphocytes (Luce et al. 2009). Both, MHC class I upregulation and CD95 may enhance tumor

cell recognition and killing after radiation. T cells require co-stimulation to become fully

activated, Seo et al.(Seo et al. 1999) demonstrated that CD80 was upregulated on B cell

lymphoma and the same was demonstrated on myeloid leukemia cells after irradiation (A. Morel

et al. 1998). Thus, radiation can enhance co-stimulation and in this way it may prevent T cell

anergy. Adhesion molecules, including intercellular adhesion molecule-1 (ICAM-1) (Hallahan,

Kuchibhotla, and Wyble 1996; Garnett et al. 2004) and vascular cell adhesion molecule-1

(VCAM-1) (A. A. Lugade et al. 2008; A. a Lugade et al. 2005) have been shown to be up-

regulated on tumour cells or in the tumour vasculature after irradiation. This increased

expression of adhesion molecules enhance the migration of lymphocytes into the tumor and

increase the interaction between immune cells (Carlos 2001). Radiation also greatly increase the

level of tumour-associated antigens (TAAs) released in the tumor microenvironment (Garnett et

al. 2004; Hareyama et al. 1991). The increased availability of released TAAs for uptake by

42

circulating DCs can result in tumor-specific immune attack. Zhang et al. (B. Zhang et al. 2007)

demonstrated that irradiation tumor with low expression of antigen, caused a significant release

of antigen and that was enough to cause tumor cell killing by cytotoxic T lymphocytes. In

summary, radiation causes increased surface expression of MHC class I and CD95 on tumor

cells, increased expression of adhesion molecules in vessels, expression of costimulatory

molecules and increase levels of TAAs. All these effects increase tumor cell recognition by

effector cells, promote lymphocyte infiltration, prevent T cell anergy, and render tumor cells

more susceptible to CTL killing.

1.4 The immune response to surgery

1.4.1 The Surgical Stress response

Surgery or trauma lead to the “surgical stress” response. This response is a combination of

metabolic, immunologic and hematologic changes occurring after injury or trauma. The severity

of the response is proportional to the magnitude of injury and reflects increased demand of organ

function (Kehlet 1997). It is generally believed that the final outcome of the surgical stress

response is postoperative immunosuppression. This is particularly important in patients with

cancer since immunosuppression may promote tumor proliferation and metastasis (Dąbrowska

and Słotwiński 2014).

Following surgical trauma, the nervous system activates the stress response by sending impulses

to the hypothalamus via afferent nerves from the injury site. This in turn stimulates the

hypothalamic-pituitary-adrenal (HPA) axis and sympathetic nervous system. The final outcome

43

is the elevation of cortisol, glucagon, catecholamines, aldosterone, vasopressin and a host of

inflammatory cytokines in an effort to provide the body with energy, retain fluid and salt and

maintain cardiovascular homeostasis. However, this state can result in outcomes such as

hyperglycemia, cardiovascular instability and immunosuppression (Desborough 2000).

The hypersecretion of cortisol and catecholamines, due to surgical stress, has both anti-

inflammatory and immunosuppressant effects (Ogawa et al. 2000). Cortisol shift the Th1/Th2

balance by interfering with the production of Type 1 cytokines and increasing the synthesis of

Type 2 cytokines, by acting directly on CD4+ T cells and indirectly by inhibiting IL-12

production by monocytes (Marik and Flemmer 2012). Also, cortisol induces downregulation of

IL-12 receptors in T cells and NK cells (Ilia J. Elenkov 2004). IL-12 is involved in the

differentiation of naïve T cells into Th1 cells, and stimulates the production of IFN-γ by T cells

while inhibiting IL-4 synthesis. Catecholamines, through the stimulation of β2-adrenergic

receptors on macrophages/monocytes, contribute to the shift in Th1/Th2 balance by inhibiting

IL-12 synthesis and enhancing the production of IL-10 (Mizuno et al. 2005). Prostaglandin E2

(PGE2) which is produced by macrophages after tissue injury further increases Th2 cytokine

production while reducing Th2 cytokines(Lorne et al. 2008). Cortisol and PGE2 act

synergistically and cause immunosuppression after trauma. Furthermore, cortisol impedes

aggregation of macrophages and neutrophils at site of injury, and decreases phagocytosis. In

addition, it induces apoptosis, in T lymphocytes and promotes Th2 cell dominance (Jameson et

al. 1997). Regulatory T cells might also be involved in the immune suppression after surgery, as

demonstrated by MacConmara (MacConmara et al. 2006), who demonstrated significant increase

in Tregs by day 7 after trauma, these regulatory T cells were a significant source of Th2

cytokines in this cohort of patients. Furthermore, Ochoa et al. (Bryk et al. 2010; Zhu, Herrera,

and Ochoa 2010) demonstrated that arginase 1 (ARG1) is induced in myeloid-derived suppressor

44

cells (MDSCs) after surgery and trauma. Th2 cytokines, catecholamines and PGE2 induce the

expression of MDSC and act synergistically to increase the expression of ARG1 in these cells

(Marik and Flemmer 2012). This leads to a state of arginine deficiency, which is required for

lymphocyte proliferation, causing further immune depression. Moreover, the combination of

surgery, neuroendocrine response and analgesics, especially opioids, depress natural killer cell

activity (Beilin et al. 2003; Melamed et al. 2003). NK cells are an important component of the

innate immune system and play a crucial role in identifying and lysing tumor cells.

In summary, the activation of the surgical stress response causes elevation of cortisol,

catecholamines and PGE2 that result in a switch in the Th1/Th2 balance, promoting release of

Th2 cytokines. Th2 cytokines in turn induce the expression of ARG1, which depletes cellular

arginine and together with an increase in Tregs result in further impairment of T cell proliferative

responses.

1.4.2 Post-surgical cytokine cascades

The changes after surgery are related to the extent of surgical trauma and neuroendocrine stress

response (Pirttikangas et al. 1995). There is a balance between the release of pro and anti-

inflammatory cytokines. Multiple regulator mechanisms exist to maintain homeostasis and avoid

an unbalanced inflammatory state. Following surgical trauma there is an early proinflammatory

immune response, also called systemic inflammatory response syndrome (SIRS) and a late

adaptive response or compensatory anti-inflammatory response syndrome (CARS) (Kimura et al.

2010). After tissue damage, the initial proinflammatory response is mediated primarily by the

cells of innate immune system, whereby phagocytic and endothelial cells release IL-1β and TNF-

45

α. These cytokines are necessary mediators that direct the inflammatory response to sites of

infection and injury, and play an essential role in promoting wound healing and maintaining

homeostasis (Lin, Calvano, and Lowry 2000). IL-1 and TNF-α leads to cleavage of Iκβ and the

subsequent activation of NF-κβ triggering the synthesis of other proinflammatory cytokines. IL-6

and IFN-γ are the primary proinflammatory mediators induced by NF-κβ. IL-6 exerts both pro

and anti-inflammatory effects and has been shown to correlate with the duration of surgery and

the extent of injury. (Menger and Vollmar 2004). IL-6 induces the production of acute phase

reactants such as C-reactive protein and procalcitonin. Also, IL-6 plays a major role in the

proliferation of polymorphonuclear (PMN) progenitors in the bone marrow and later in the

function of mature PMNs (Botha et al. 1995). Lastly, IL-6 act as an anti-inflammatory cytokine

and helps controlling local or systemic acute inflammatory responses (Xing et al. 1998). IL-6

exerts anti-inflammatory properties during injury by attenuating TNF-α and IL-1 activity,

furthermore, it promotes the release of sTNFRs and IL-1ra (Lin, Calvano, and Lowry 2000).

The early proinflammatory response following surgery is a result of a predominance of the Th1

cytokines (IL-2, IL-12, and IFN-γ). However, the increased surgical stress release of

glucocorticoids, catecholamines and acute phase reactants often results in a shift towards the

anti-inflammatory Th2 predominance (cytokines IL-4, IL-5, IL-6, IL-10, and IL-13) later in the

postsurgical period with consequential depressed cellular immunity (Menger and Vollmar 2004;

Lin, Calvano, and Lowry 2000).

46

1.4.3 Postoperative tumor progression

Surgery is the most successful therapy when treating patients with solid tumors (Demicheli et al.

2008). However, occasionally radical surgery accelerates growth and dissemination of residual

malignant cells (Kal, Struikmans, and Barten-van Rijbroek 2008). In the wound bed, cells that

normally divide infrequently, are induced to proliferate rapidly, epithelial cells and stromal cells

migrate and new blood vessels are recruited. Thus, a wound response would appear to provide a

highly favourable milieu for cancer progression (Hofer et al. 1999). Furthermore, enhanced

tumor progression following surgery is thought to be largely mediate by NK cell suppression

(Colacchio, Yeager, and Hildebrandt 1994; Shamgar Ben-Eliyahu, Page, and Schleifer 2007; S

Ben-Eliyahu et al. 1999). Animal studies indicate that the majority of anesthetics have a

profound suppressive effect on NK cells and some of them are associated with increased

metastases (Welden et al. 2009). A balanced Th1/Th2 ratio is important for anticancer immunity

(Melamed et al. 2003). The characteristic change in balance towards an Th2 type immune

response after surgery is associated with depressed cellular immunity and tumor surveillance

(Wada et al. 2007). Finally, surgical manipulation may result in shedding of tumor cells and

release of growth and angiogenic factors (VEGF) (Hormbrey et al. 2003), thus promoting

metastasis and tumor growth. In summary, a favourable milieu for proliferation, the anti-

inflammatory response following surgery with the dominant Th2 response, and NK cell

suppression contribute to tumor progression and metastasis after surgical trauma.

1.5 Summary

47

MPM is a cancer of the pleura associated with the inhalation of asbestos fibers. Due to a long

incubation time between exposure and onset of disease, the incidence of disease in North

America has increased by 65 percent over the past two decades worldwide and is projected to

continue to increase until at least the year 2020. Moreover, the continued use in the developing

world suggests that MPM will continue to rise worldwide for the foreseeable future. Median

survival of MPM patients without treatment is 4 to 9 months. Non-resectable MPM is treated

with a pemetrexed and platinum based chemotherapy regimen that increases median survival

from 9 to 12 months in epithelial disease. Conventional therapies to MPM offer little

improvement in survival, thus newer therapeutics are needed.

The group of Dr. De Perrot and Dr. Cho has developed a new approach consisting in

hypofractionated radiation followed by Extrapleural Pneumonectomy which differs from

previous approaches in which radiation is given after surgical resection. A feasibility study of

Surgery for Mesothelioma After Radiation Therapy (SMART) has demonstrated that this

procedure results in a significant increase in 3-year survival in patients with epithelial disease

from 53% to 84% compared to previous treatment modalities. The authors discussed, based on

accumulating evidence, that high-dose hypofractionated radiation is able to stimulate the immune

system in addition to mediating direct tumor killing. Cytotoxic T lymphocytes circulate through

the body and are able to recognize and eradicate tumor cells. However, a strong inhibitory tumor

microenvironment renders T cells unable to inhibit tumor growth. Radiation has been shown to

reverse this immunosuppressive environment and promote a strong inflammatory response which

may shift the balance towards an anti-tumor response. In the SMART approach the pro

inflammatory effect of radiation together with the removal of the immunosuppressive tumor may

have contributed to the beneficial effects seen in patients. This pro inflammatory effect of

radiation may open the door to newer therapeutic approaches against cancer such as combination

48

with immunotherapy. In this project, we investigated the effect of local radiation therapy in

combination with surgery and immunotherapy in a mouse model of mesothelioma. We analyzed

the immune response after radiation and the role of CD8+ T cells. Finally we assessed the role of

lymphocytes in the generation of a protective immunological memory in our mouse model. The

following chapter will outline the hypothesis and aims of this study.

49

2 Hypothesis and Aims

Radiation therapy (RT) is an important modality in the treatment of mesothelioma and many

other tumors. Until recently RT was used based on the ability to eradicate cancer cells by means

of its cytotoxic effect [1]. In regards of its interaction with the immune system, it was considered

an immune attenuator. If we consider the effect of radiation to any particular cell in isolation, it

will be almost always detrimental. However, if we account for the interaction between tumor cell

death, enhanced antigen expression on tumor cells, and inflammatory signals from the irradiated

tissue which affects lymphocytes and dendritic cells (DC), we could have a beneficial effect on

the immune system.

The goal of this study was to investigate the effect of local radiation therapy of a tumor in

combination with surgery and immunotherapy in the context of mesothelioma in a mouse model.

The goal of this translational research projects was to use the findings of this study to understand

and improve the treatment approach of MPM.

The objectives of this study were threefold: 1) to develop a mouse model of MPM to study the

effect of local radiation therapy and surgery; 2) to evaluate CD8+ T cell immune response on

tumors after radiation therapy; 3) to investigate the effect of radiotherapy combined with

immunotherapy, specifically the antibody anti CTLA-4.

For the first model a subcutaneous tumor is established in the right flank of the mouse. Tumor

growth is monitored and treated when the tumor reaches a threshold area. The tumor is irradiated

50

and/or removed surgically. Being a subcutaneous model it is easy to follow tumor growth and

evaluate the effect of different treatments.

I propose combination therapy with hypofractionated local radiation and surgery will have better

tumor control than either therapy alone. For the second objective, I postulate activation of the

immune system will play a role and there will be more CD8+ T cell tumor infiltration in the

group receiving radiation therapy either alone or in combination. CD8+ T cell infiltration will

correlate with tumor control and will provide treated animals with immunological protective

memory. This immunological memory will protect mice when rechallenged with the same tumor

and depletion of lymphocytes will make them susceptible to rechallenge.

For the last objective, we will examine immunotherapy in the mouse model to further support

the role of immune activation after local hypofractionated radiation. Tumor bearing mice will be

treated with anti CTLA-4 antibody alone or in combination with radiation, surgery or both. It is

hypothesized that anti CTLA-4 will have a synergistic effect when combined with radiation

therapy. The hypothesis that radiation induces an immunogenic cell death and promotes

recruitment and function of T cells within the tumor microenvironment supports the idea that a

tumor can be converted into and in situ vaccine. This effect of radiation might be relevant and

provide a synergistic effect when combined with immune checkpoint inhibitors such as anti

CTLA-4 antibody.

51

3 Materials and Methods

3.1 Murine Cell lines

AB12 and AE17 malignant pleural mesothelioma cell lines were both derived from an asbestos-

induced tumor in a BALB/c and C57BL/6 mouse, respectively. AB12 was kindly donated by Dr.

Jay Kolls, University of Pittsburgh, Pittsburgh, PA, in 2008. AE17 was obtained from the

European Collection of Cell Cultures. AE17-OVA was developed by stably transfecting the

parental cell line (AE17) with secretory ovalbumin (sOVA).(Jackaman et al. 2003) The cell line

was kindly provided by Dr. Steven Albelda, University of Pennsylvania, Philadelphia, PA, and

Dr. Delia Nelson, University of Western Australia, Crawley, WA, Australia.

AB12 and AE17 were grown in RPMI 1640 culture media (Life Technologies Inc., Burlington

ON, CAN) supplemented with 10% heat inactivated fetal bovine serum (Life Technologies Inc.,

Burlington ON, CAN), 2 mM L-glutamine, 100 units/mL penicillin, 100 μg/mL streptomycin

and non-essential amino acids. The transfected cell line AE17-OVA was maintained in the same

medium supplemented with 400 μg/L neomycin analog G418 (geneticin; Invitrogen). Cells were

plated in tissue-culture coated flasks (BD Biosciences Canada, Mississauga, ON).Additionally,

cultures were grown in a 37oC and 5% CO2 environment and passaged when 70% confluent. For

passage, cells were trypsinized using 0.25% trypsin and split 1 in 10.

52

3.2 Mice

Eight to twelve week old C57BL/6 and BALB/c wild type mice were purchased from the

Jackson Laboratory (Maine, USA). All mice were housed at the Toronto Medical Discovery

Tower’s Animal Resource Centre under pathogen-free conditions in accordance with

institutional and national animal care and ethics protocols.

3.3 In vivo Tumor Growth experiments

Mice were injected subcutaneously in the right flank with 1x106 AB12 cells, AE17 or AE17-

OVA cells in 100μL of PBS at day 0. For rechallenge experiments cells were injected s.c. in the

left flank. After removing fur and cleaning the skin, injections were made with a syringe and 25-

27G needles. Tumor growth and weight was monitored 3 days a week. Tumor dimensions were

measured using microcallipers. Mice were regularly checked and sacrificed when tumor

dimension reached 150mm2 or showed signs of ulceration as per institutional ethics protocols.

3.4 Local Radiation Therapy

For all experiments involving local radiation therapy, eight to twelve week old BALB/c mice or

C57BL/6 mice were injected subcutaneously in the right flank with 1x106 AB12 cells or AE17-

OVA cells in 100μL of PBS, respectively at day 0. On radiation day, tumors were either sham-

irradiated or irradiated with a total dose of 15 Gy over 3 days. Radiation was given using the X-

Rad 225Cx small animal image-guided irradiator (Precision X-Ray, Branford CT, USA). The

53

irradiator has a 225kVp x-ray tube (Varian Associates, Palo Alto, CA) and a flat-panel silicon

detector mounted on a 360o rotation C-arm gantry.(Moretti 2010) The automated stage is

movable on the x, y and z axis. It is all housed in a self-shielded cabinet and is remotely

controlled by a computer (Dell Precision 690, Intel Xeon CPU running Windows XP). The mean

targeting displacement error is approximately ≤0.1mm in the x-y-z planes. Radiation was given

to mice under isoflurane anesthesia and positioned on the irradiation stage. To initially visualize

the animal and the tumor fluoroscopic mode was used. To target precisely the tumor a scout

cone-beam computed tomography was done at a 40kVp tube potential and 0.5mA current. The

tomography was then reconstructed at a 0.4mm voxel size (Moretti 2010). The beam source was

collimated to either a 1.5 cm or 2 cm diameter circular field. To confirm the area to be irradiated

the tumor was then visualized under fluoroscopic imaging with the collimator in place,

immediately prior to delivery of treatment. Radiation was delivered at a tube potential of 225kVp

and a 13 mA current for a dose rate of 3.02 Gy/min. The daily dose was given from 2 angles,

half from above and half from below. Total dose was given in divided fractions over 3 days

according to treatment protocols. After radiation mice were placed back in their cages and

housing facilities.

3.5 Surgical Resection of Subcutaneous Tumors

Mice with flank tumors under general anesthesia with isoflurane were shaved and cleaned with

isopropanol. Tear gel was applied on both eyes and a heating lamp was used to prevent

hypothermia. Skin around the tumor was infiltrated with Marcaine (bupivacaine 0.25%) prior to

incision. Two different approaches were used depending on the experiment. We named the first

54

approach, “Blunt Surgery”, and the second and more aggressive approach, the “Radical

Surgery”. For both approaches, a 1 cm incision was made adjacent to the tumor. Blunt Surgery

was achieved using careful blunt dissection. All macroscopic tumor was removed but no skin or

surrounding healthy-looking tissue (Figure 3)

Figure 3 Partial resection of a subcutaneous tumor with blunt dissection. Under general

anesthesia the animal was positioned in the lateral decubitus position with the tumor exposed and

removal of the tumor was performed. A) After infiltration around the tumor with bupivacaine a 1

cm incision was made along the tumor. B) With blunt dissection the tumor was removed. C)

Complete macroscopic resection of the tumor was done but no skin or surrounding tissue. D) The

wound was closed with 6-0 prolene.

A

C

B

D

55

Radical Surgery was performed removing skin on top of the tumor and a 0.5 cm margin of

healthy skin around the tumor. All macroscopic tumor was removed including vascular supply

and 0.5 cm margin of healthy-looking subcutaneous tissue around the tumor. Sterile prolene 6-0

or 5-0 sutures were used to close wounds. Marcaine was administered immediately after closing

the wound along the suture for postoperative analgesia and the mice were observed until

complete recovery. Mice were then monitored 6hrs, 24hrs and 48 hrs after surgery and

meloxicam 1mg/kg SC was given for postoperative analgesia in case of mild or moderate pain.

3.6 Combination therapy with LRT and Surgery

For the experiments involving combination therapy with LRT and Surgery tumors were

inoculated in the right flank. After 7 to 10 days when all tumors were larger than 6 mm2, mice

were treated with LRT for 3 consecutive days as described before and depending of the

experiment, 24hrs, 5 days or 7 days after, surgery was performed.

3.7 In vivo depletion of CD4+ and CD8+ specific T cells

Anti-CD4 MAb from rat GK 1.5 hybridoma or anti-CD8 Mab from 2.43 rat hybridoma (Bio X

Cell, West Lebanon, NH, USA) were diluted to a final concentration of 1 mg/mL with PBS or

2mg/ml for double depletion. Intraperitoneal injections for 3 consecutive days with 0.2 ml (0.2

mg) of purified Mab were performed. For double depletion the total volume injected was 0.2 ml,

consisting of 0.1 ml of each Mab at a concentration of 2mg/mL. By day 6, peripheral lymphoid

56

organs were depleted and one control was sacrificed to assess satisfactory depletion (>95%). The

depleted condition was maintained with 0.2 mg injections of MAb every 3 days.

3.8 Anti-CTLA-4 therapy

Mouse monoclonal antibody (9D9) to CTLA-4 (Bio X Cell, West Lebanon, NH, USA) was

diluted to a final concentration of 1mg/mL with PBS and kept at 4°C until further use.

Intraperitoneal injections were made with 0.2 mL (0.2 mg) of the purified antibody every 3 days

for the length of the specified treatment.

3.9 Blood Collection

Blood was collected by tail venipuncture or heart puncture using heparinized capillary blood

collection tubes (Fischer Scientific Co., Toronto, ON). Red blood cells were lysed with ACK

lysing buffer (Life Technologies Inc. Burlington, ON). Finally, remaining cells were washed

twice with phosphate buffered saline, pH 7.4 (Life Technologies Inc., Burlington ON, CAN).

3.10 Tumor Digestion

Tumors were removed and placed in 15 mL conical tubes filled RPMI 1640 culture media and

stored on ice until further use. To prepare for dissociation the tissue was chopped to small pieces

(approx. 2 mm) and transferred to 15 mL conical tubes containing digestion media consisting in

57

RPMI 1640, DNAse (Roche 10104159001) and Liberase TM (Roche Diagnostics, Laval,

Quebec, Canada). Tubes were placed in a shaking water bath for 30 minutes and when the pieces

were soft and malleable the solution was filtered and mashed through a 70 μm cell strainer. Cells

were then washed with PBS and remaining cells were counted and viability was assessed.

3.11 Isolation of Lymphocytes from Spleens and Lymph Nodes

Spleens and inguinal and axillary lymph nodes were isolated in phosphate buffered saline (PBS)

and filtered through a 70μm cell strainer. Red blood cells were lysed with ACK lysing buffer

(150mM NH4Cl, 10mM KHCO3, 0.1mM NA2EDTA) for 10 minutes at room temperature and

washed twice with PBS after lysis. Remaining cells were counted using trypan blue exclusion

and a hemocytometer.

3.12 Flow Cytometry

Cells were resuspended in FACS buffer (PBS, 2% FBS, EDTA 5mM) and stained for 30 minutes

at 4 C with α-CD16/CD32 Fc block (BD, Pharmingen; San Diego, CA, USA), and a combination

of the following mouse specific antibodies: CD3, CD4, CD8, CD44, CD45, CD69, CD137 (4-

1BB), TIM3, PD-1, ICOS. Cells stained with tetramer were incubated for 30 min with the Class I

H-2Kb SIINFEKL tetramer prior to surface staining. All samples were then washed twice with

FACS buffer and analyzed immediately. FACS analysis was conducted using a BD LSR II flow

cytometer (BD Biosciences, Mississauga ON, CAN). Analysis was performed using FlowJo

V10 (FlowJo LLC, Ashland, USA) software.

58

3.13 Immunofluorescence

Frozen tissue samples on slides were fixed with cold acetone for 10 minutes. Paraffin embedded

samples were deparaffinised with Xylene, 100% ethanol, 95% ethanol, and 70% ethanol

respectively and antigen retrieval was performed by immersing samples in 100°C citrate buffer

for 20 minutes. Samples were blocked with 5% BSA in Tris-buffered saline for one hour before

the addition of primary Ab. After incubating overnight at 4°C, sections were washed in

TBS+0.2% Tween 20. Slides were subsequently incubated for 1 hour at room temperature with

the appropriate fluorescently labelled secondary Ab. Slides were further washed with

TBS+0.2% Tween 20 before adding mounting media with DAPI nuclear stain. Cover slips were

placed on top and sealed with nail polish.

Fluorescently labelled cells or tissues were visualized with the WaveFX (Quorum Technologies

Inc, Guelph ON, CAN) confocal microscope system. Pictures were analyzed using ImageJ

V1.47 (National Institute of Health, USA). Corrected total cell fluorescence (CTCF) was

calculated by the formula CTCF = Integrated Density – (Area of cell x mean fluorescence of

background reading).

3.14 Immunohistochemistry

Tumors were snap frozen in liquid nitrogen and kept at –80oC on dry ice until staining. Frozen

samples were sliced into 5µm thick sections using a microtome. Frozen sections were fixed with

1% paraformaldehyde for 1 hour before staining. Sections were blocked with serum (5% BSA in

Tris-buffered saline) for one hour before the addition of primary anti-CD3, or CD8 antibody at a

59

1:100 dilution. After incubating overnight at 4oC, sections were washed in TBS+0.2% Tween

20. Slides were subsequently incubated for 1 hour at room temperature with anti-rabbit-HRP

secondary Ab kit (Vector Laboratories Inc., Burlington ON, CAN). After washing, the HRP

substrate DAB (3, 3-diaminobenzidine) (Vector Laboratories Inc., Burlington ON, CAN) was

added to each slide at 100 µL per section. Sections were then counterstained with hematoxylin,

dehydrated, and mounted with mounting media (Fischer Scientific, Ottawa ON, CAN).

Immunostained slides were imaged at 200x magnification with the Aperio ImageScope digital

scanner and visualized with Aperio ImageScope Viewer version 12.1 (Vista, CA, USA).

Scanning was provided by the Advanced Optical Microscopy Facility (Toronto ON, CAN)

3.15 Ovalbumin ELISA

AE17-OVA and AE17 cell culture supernatants were collected 3 days after seeding cells. Cells

were then trypsinized and washed twice with PBS. For cell lysates, cells were collected by

centrifugation, 5 min at 1000xG. Cells were then subjected to ultrasonication for 4 cycles on ice.

Cell lysates were collected by centrifugation at 1500xG for 10 minutes at 4 °C to remove cellular

debris. Cell lysate or media was placed in wells coated with a biotin-conjugated antibody

specific to OVA from an ELISA kit (Biomatik corporation, Cambridge, ON). Samples were left

for 2 hours to bind anti-Ova antibodies. Avidin conjugated to Horseradish Peroxidase (HRP) was

then added to each well and incubated for one hour. Finally TMB substrate solution was added

and those wells containing OVA, biotin-conjugated antibody and enzyme-conjugated Avidin

exhibited a change in color. Reaction was terminated by the addition of sulphuric acid.

60

Concentrations were determined by four-parameter logistic test using a standard curve. Samples

were measured in duplicate.

3.16 Statistical Analysis

Statistical analysis was performed with GraphPad Prism 5 (GraphPad Inc, La Jolla CA, USA).

More than 2 groups were compared using one-way ANOVA analysis. Student’s T-test was used

to analyze two groups. A P-value of less than 0.05 was considered statistically significant.

Results have been presented as Mean ± Standard Error of the Mean (SEM). * indicates p<0.05,

** indicates p<0.01, and *** indicates p<0.001 in all results figures.

61

4 Results

4.1 Development of a Mouse Model of Malignant Mesothelioma

To develop a mouse model of mesothelioma, BALB/c mice and C57BL/6 mice were inoculated

with the cell lines AB12 and AE17-OVA respectively.

4.1.1 Local Radiation Therapy, Right Flank Model

To determine the radiation sensitivity of tumors in vivo, tumor irradiation with 4 different doses

was conducted. Initially experiments were done using BALB/c mice and the cell line AB12

following a previous model used by our group. The tumor was inoculated subcutaneously in the

right flank with 1x106 AB12 cells. The right flank was chosen because it is accessible for

consistent caliper measurements and readily accessible for surgical resection of the tumors.

Tumors were locally irradiated with targeted x-rays starting 7 days after tumor inoculation

(Error! Reference source not found.). On the first day of treatment, mice were randomized into

the following groups: 1) no treatment, 2) 15Gy over 3 days (5Gy x 3), 3) 22.5Gy over 3 days

(7.5Gy x 3), 4) 30Gy over 3 days (10Gy x 3) and 5) 22.5Gy in a single dose. Tumor growth was

followed every 3 days with caliper measurement before and after treatment until they reached a

humane endpoint, at that point mice were sacrificed. Tumor size is expressed as tumor area in

squared millimeters using the longest length and the perpendicular width (Length x width).

62

Figure 4. Schematic of radiation treatment in tumor bearing mice. Tumor was inoculated on

day 0. In the treated group, Local Radiation Therapy (LRT) started on day 7. Depending on the

group, radiation was give on 3 consecutive days or on a single dose. Tumor was monitored every

3 days with caliper measurements.

Untreated mice showed the most rapid tumor growth and greatest tumor area. After 22 days

tumor growth decelerated (results not shown) and mice were in distress, at that time point mice

were sacrificed. The tumor growth in untreated mice could be described as a sigmoidal curve.

Radiation with 30Gy (10x3) and 22.5G in one fraction exhibited an excellent response to

radiation compared to no treatment. Tumor growth stopped for 10 days after treatment started

and tumor area was significantly lower on day 16, 19, and 22 compared to no treatment (

Figure 5). However, irradiated mice showed signs of distress and lost 10 to 15% of total body

weight during the first 2 weeks after treatment (data not shown). Radiation with 15Gy (5x3) and

63

22.5Gy (7.5x3) displayed a good response, with tumor growth deceleration for 7 to 10 days after

treatment. Tumor area was significantly lower compared to untreated mice at days 16, 19 and 22.

There was no significant difference among LRT treatment groups.

Overall, tumor growth slowed down in mice treated with local radiation therapy (LRT) compared

to untreated controls for at least 6 days in all cases. The difference was significant for all groups

receiving 15Gy and 30Gy at in 3 fractions. After approximately 7 to 10 days post-radiation,

tumor growth accelerated in all groups. This result confirms that the tumor model is sensitive to

LRT. For the following experiments 15Gy was used, consistent with others in our group and

because it was the lowest radiation dose with good effect in tumor growth without accompanied

weight loss or mouse distress.

64

A

B

Figure 5 Increasing doses of Local Radiation Therapy and its effect on tumor growth. A)

Tumor area is represented as the mean of 5 mice. All 4 LRT doses showed an excellent response,

with tumor growth stable for at least 7 days B) Tumor is significantly smaller in the 4 LRT

groups compared to untreated mice on days 16, 19 and 22. Values shown are the mean ± SEM of

5 mice per time-point. ** < 0.05, n.s not significant comparing treated groups to untreated.

65

4.1.2 Combination therapy with LRT and Surgery

To evaluate the different treatment modalities in the BALB/c mouse model and to assess whether

or not LRT combined with surgery had a benefit, the different treatment modalities were

compared. In this experiment blunt surgery was used to remove tumors. Mice were randomized

the first day of LRT into the following groups: 1) No treatment, 2) Local Radiation Therapy

15Gy (5x3), 3) Surgery, 4) LRT and Surgery. In the LRT and Surgery group, blunt surgery was

performed 5 days after completion of LRT (Figure 6). For statistical analysis the day of surgical

removal of the tumor is day 0 for the groups treated with blunt surgery, otherwise day 0 is the

day of the inoculation of the tumor.

Mice treated with surgery alone had tumor recurrence as early as 48 hours after surgery. Tumor

growth rate after tumor resection was more rapid than untreated tumors and tumor size was

significantly larger than untreated mice at days 17 and 24. In the group treated with LRT alone

tumor growth slowed down significantly compared to untreated mice. Tumor area was

significantly smaller at day 24 and 28. Finally, in the combination group, tumor growth slowed

down after LRT and prior to surgical removal. Tumor growth after surgery was not observed

until at least 7 days later. Tumor size was significantly smaller compared to untreated tumors on

days 17 and 28 (n=5, p=0.035; n=5, p=0.0415) (Figure 7).

Overall, tumor growth after blunt surgery alone was accelerated compared to untreated mice.

Conversely, tumor growth slowed down in the mice treated with LRT alone or LRT and blunt

surgery. This suggest blunt surgery alone may have negative effects in the treatment of tumors

and is not a good therapeutic approach in this model. However, when LRT was given prior to

66

blunt surgery tumor growth slowed down significantly postoperatively and abrogated the

negative effects of surgery.

Figure 6 Schematic of LRT and Surgery in AB12 tumor bearing mice. Tumor was

inoculated on day 0 and treatment started on day 12. In the combination treatment group, blunt

surgery was performed on day 19, 5 days after completion of LRT. Tumor was monitored every

3 days with caliper measurements.

67

Figure 7. The effect of combination therapy with LRT and Surgery. LRT alone or in

combination with blunt surgery slows down tumor growth. However, blunt surgery alone accelerates

tumor growth compared to no treatment. A) Untreated group B) Blunt surgery alone day 12. C) LRT

alone day 12 D) Combination group, LRT at day 12 and blunt surgery 5 days after. E) Groups means

showing a significant negative effect of surgery alone and a significant beneficial effect on the groups

treated with LRT F) Significant difference at days 17 and 24 in the surgery group compared to no

treatment and at days 24 and 28 in the LRT treated groups compared to untreated. In E) and F) the day of

surgical removal of the tumor is day 0 for the groups treated with blunt surgery, otherwise day 0 is the

day of the inoculation of the tumor. Values shown in E) and F) are the mean ± SEM of 5 mice per time-

point. ** < 0.05 compared to untreated, §<0.05 compared to surgery.

68

4.2 T cells infiltrate tumors after LRT

4.2.1 Tumor infiltrating CD8+ T Cells in Untreated and Radiated Mouse

Tumor Tissue

To evaluate if the immune system was involved in the effect of LRT on tumor growth we

analyzed CD8+ T cell tumor infiltration after LRT. CD8+ T Cells are responsible for mediating

antigen-specific tumor cell killing. Staining for CD8+ T Cells was performed on untreated and

radiated tumors to study the effect of radiation in terms of CD8+ T Cell recruitment. The

recruitment of the CD8+ T Cell immune response was measured by immunofluorescent staining

of tumor tissue for CD3+CD8+ cells. Tumor samples from 15Gy radiated and untreated mice

were taken 2, 7, and 12 days after LRT for the pathology analysis and on day 7 for FACS

analysis. Tumors in the treated group were visibly smaller compared to untreated tumors at days

7 and 12 after LRT. To confirm results, analysis was done with flow cytometry of the treated or

untreated tumors, stained with conjugated antibodies specific for CD45, CD3 and CD8. For

FACS analysis 3 tumor samples were pooled and analyzed as a single sample.

The number of CD3+CD8 T cells on the slides was quantified at each time point by averaging

the cell counts of 5 random fields (Figure 8a). The count of double positive cells 2 days after

LRT was not significantly different (18.40 ± 4.739 vs. 15.20 ± 2.818 cells, p= 0.5775). However,

7 days after LRT the count was significantly higher in the LRT group compared to untreated

tumor (32.20 ± 8.225 vs 10.60 ± 3.076 cells, p=0.0398). Similarly, 12 days after LRT, treated

tumors showed higher counts compared to untreated (55.60 ± 13.07 vs. 1.600 ± 0.5099 cells,

p=0.0033) (Figure 8b). Peak staining occurred 12 days after initiating radiation. It was also

noted that the number of double positive cells decreased in the untreated tumor over time (days 7

69

and 12) and as the tumor grew bigger. This was in contrast to what was observed in the treated

tumor. We confirmed these results with FACS, where we observed 5.19% CD45+ CD3+CD8+

cells infiltrating the treated tumor compared to only 1.03% in the untreated tumor 7 days after

the first day of radiation (Figure 8c).

In summary, treated tumors showed a significant increase in the number of infiltrating

CD3+CD8+ cells on days 7 and 12 after LRT. This suggest LRT recruits CD3+CD8+ T cells

into the tumor.

70

C

Figure 8. CD3+CD8+ cell tumor infiltration after LRT compared to untreated tumors.

Immunofluorescent staining of tumor 2, 7 and 12 days after LRT, compared to untreated tumor

controls. A) Images show DAPI (405nm), CD3 (488 nm) and CD8 (633nm) merged staining. B)

Average cell count of 5 random x200 magnified fields. *<0.05, **<0.005 C) FACS analysis of

the treated and untreated tumor 7 days after the first day of radiation. Doublets and dead cells

were excluded before gating on CD45/CD3. FACS confirms the increase number of CD3+CD8

cell infiltrating the tumor after LRT compared to untreated control. The graph represent 4 tumors

pooled into a single tube.

71

4.2.2 A Large Proportion of Tumor Infiltrating Lymphocytes are OVA-

specific

To examine whether or not the increased number of tumor infiltrating CD8 T cells in the treated

tumor were tumor-specific, tumor infiltrating cells were stained using H-2Kb tetramers

containing the OVA protein-derived peptide SIINFEKL. For this experiment C57BL/6 mice and

the cell line AE17-OVA were used. Ten days after LRT treated and untreated tumors were

excised, and analyzed with FACS.

After gating for live cells, CD3 and CD8 double positive cells were identified. Out of the

CD3+CD8+ double positive population, the proportion of CD44+ and SIINFEKL tetramer cells

were identified (Figure 9). Radiated tumors showed greater proportion of tetramer specific

CD8+ T cells compared to untreated tumors. This difference was close to significance comparing

4 tumors in each group (30.60 ± 6.785 n=4 vs 14.99 ± 2.554 n=4, p=0.07).

The result of this experiment gives further evidence that LRT promotes recruitment of

lymphocytes into the tumor. About 30% of the recruited lymphocytes are specific for the OVA

derived peptide SIINFEKL in the treated tumor compared to only 14.99% in the untreated group.

Together with the previous experiment this suggests radiation of a tumor with 15Gy in 3

fractions in our model stimulates recruitment of CD8+ T cells to tumor tissue. A high proportion

of the CD8+ T cells are specific against our tumor antigen OVA.

72

A B

Figure 9. CD8+ lymphocytes infiltrating AE17-OVA tumor are OVA specific. A)

Representative flow cytometry graph gated on CD3+, CD8+. There is greater proportion (50.8%)

of CD44+ Tetramer+ double positive cells in the radiated group compared to untreated tumor

(21.4%). B) Graph comparing proportion of tumor specific CD8+ T cells in radiated and

untreated tumor.

73

4.2.3 Expression of 4-1BB and PD-1 by Tumor Infiltrating Cells

To assess the phenotype of tumor infiltrating lymphocytes following LRT we assessed the

expression of the inhibitory receptor, PD-1 and the activation marker 4-1BB on tumor specific

CD8 T cells. C57BL/6 mice and the cell line AE17-OVA were used. LRT treated tumor and

untreated controls were analyzed 3 and 10 days after completion of LRT. Flow cytometric

analysis was utilized to quantify the frequency of CD8+ Tetramer+ 4-1BB+ or CD8+ Tetramer+

PD-1+.

The early activation marker 4-1BB was significantly upregulated in the treated tumors 3 days

following radiation compared to untreated controls (8.932±2.521 n=5 vs 1.840 ±0.280 n=5,

p=0.0233) but 9 days later there was no difference (4.938±3.393 n=4 vs 4.1418±1.078 n=4,

p=0.0831) (Figure 10). In the case of PD-1, significantly fewer lymphocytes expressed the

marker at day 3 and 9 after radiation (16.13±5.453 n=4 vs 38.63±5.138 n=4, p=0.0239; and

30.45±12.39 n=4 vs 67.7±6.495 n=4, p=0.0374) (Figure 10).

In summary, radiation enhances the expression of 4-1BB on tumor specific infiltrating

lymphocytes and downregulates the expression of PD-1. This suggest that LRT may reverse T

cell exhaustion and restore T cell activation temporarily.

74

Figure 10. 4-1BB and PD-1 expression tetramer specific CD8+ T cells, 3 and 9 days after

LRT. 4-1BB upregulation 3 days after LRT and downregulation of PD-1, 3 and 9 days after

LRT. ** < 0.05, n.s not significant comparing treated groups to untreated.

4.2.4 Depletion of CD4+ T cells and CD8+ T cells partially abrogates the

effect of LRT on tumor growth

To examine whether or not CD4+ T cells and CD8+ T cells played a role on the response of

AE17-OVA tumors to radiation, C57BL/6 mice were depleted of CD4+ T cells, CD8+ T cells, or

both 1 day before LRT and throughout the length of the experiment. Mice were randomized to

75

the following groups: 1) No treatment, 2) LRT only, 3) LRT and CD4 depletion, 4) LRT and

CD8 depletion, 5) LRT and double depletion.

All groups treated with LRT showed significantly delayed tumor growth compared to the

untreated group at days 17, 20 and 24. Double depleted animals treated with LRT showed

significant greater tumor size than animals treated with LRT at days 24 and 27. In comparing

LRT only group and LRT and CD8 depleted groups there was a trend towards larger tumor area

in the CD8 depleted group at day 24 and 27. There was also a trend for smaller tumor size in

CD4 depleted animals treated with LRT compared to LRT only at days 24 and 27. Finally, CD8

depleted mice treated with LRT showed significant greater tumor size than CD4 depleted mice at

day 24 and 27 (Figure 11).

In summary, tumor size was significantly larger in double depleted animals treated with LRT

compared to mice treated with LRT only. This suggest T cells play a role in the beneficial effect

of LRT on tumors. Although not significant, there is a trend toward larger tumors in the LRT and

CD8 depletion group, and, toward smaller tumors in the LRT and CD4 depletion group

compared to the LRT only group. This suggests that the beneficial effect of LRT is partially

mediated by CD8+ T cell activity. On the other hand, CD4+ T cells may play a detrimental role

on radiated tumors, as shown by the trend towards smaller tumors when mice were depleted.

This effect is probably related to CD4+ regulatory cells activity within the tumor.

76

A

B

/

Figure 11. LRT and CD4+ CD8+ T cell depletion. Tumor growth in mice treated with LRT

and depletion of CD4+, CD8+ or both. Values shown are the mean tumor area in mm2 of 5 mice

per time point and are expressed as mean±SEM. ** < 0.05, compared to untreated; §<0.05

compared to LRT; ¶ compared to LRT + depletion of CD4+.

77

4.3 Immunological Protective Memory After LRT and Surgery

In this experiment, the goal was to investigate the role of LRT before surgical resection of

tumors in generating effective immunological memory response. C57BL/6 mice were inoculated

with AE17-OVA cells and after 9 days were randomized into the following treatment groups 1)

Surgery, 2)LRT and Surgery 24 hours later, 3)LRT and Surgery 7 days later. Radical surgery

was performed in this experiment to maximize the number of cured mice in order to rechallenge

them in the opposite flank 90 days after the initial treatment. Each group consisted of 10 mice.

All 10 mice in the surgery group were tumor free 90 days after treatment, and 9 mice in both

groups treated with LRT and surgery were tumor free. One mouse in the two groups treated with

LRT and surgery was lost during surgery due to tumor infiltration of the chest wall. After

rechallenge in the opposite flank, tumor size was significantly smaller in the group treated with

LRT and radical surgery after 7 days, compared to the other 2 groups (Surgery only and LRT and

Surgery 24hrs later). In the group treated with LRT and surgery after 7 days, and rechallenged

after 90 days, 3 out of 9 mice completely rejected the tumor. There was no rejections in the other

two groups (Figure 12).

This suggest that LRT 7 days before surgical removal of tumors contributes to generating a

protective immunological memory response. This protection was not generated in the group

treated only with radical surgery or when surgery was done one day after radiation.

78

A

B

C

Figure 12. AE17 OVA Rechallenge 90 days after treatment. A) Mice treated with radical

surgery only, LRT and surgery after 24 hrs or LRT and surgery after 7 day were rechallenged in

the opposite flank 90 days after treatment. B) Mice treated with LRT and radical surgery after 7

days grew significantly smaller tumors compared to the surgery and to LRT and surgery after 24

hrs. Values shown are the mean tumor area in mm2 of 10 mice per time point in the surgery

group and 9 mice in the other groups and are expressed as mean±SEM. ** < 0.05, compared to

LRT and surgery after 7 days.

79

4.3.1 Role of T cell on the protection against rechallenge

To investigate whether or not T cells play a role in the protection against AE17-OVA

rechallenge after LRT and surgery, C57BL/6 mice that rejected tumors after rechallenge in the

previous experiment were depleted of lymphocytes and then rechallenged again.

Animals treated and cured with LRT and surgery were rechallenged and completely rejected the

tumor. These mice were pooled from multiple previous experiments after varying periods of time

ranging from two to 14 months after rejection of the tumor and then prior to the depletion

experiment all mice were rechallenged with AE17-OVA in the left flank to assess if they still had

protective memory response against the tumor. Twenty one mice were collected and 20 rejected

the tumor for the second time (data not shown). Lastly, these 20 mice were rechallenged once

more after randomization into the following groups: 1) CD4 depletion n=6, 2) CD8 depletion

n=7, 3) Double depletion n=7. Depletion of lymphocytes started one week before AE17-OVA

cells inoculation (Figure 13a).

Double depleted mice displayed the fastest tumor growth rate, similar to mice challenged for the

first time. Tumor size was significantly larger than the other two groups starting at day 9 and at

every time point until mice were sacrificed. All CD8 depleted mice completely rejected the

tumor. CD4 depleted mice rejected 1 of 6 tumors and growth rate of the remaining 5 was

significantly slower than double depleted animal but also significantly faster than CD8 depleted

mice. Tumor size in CD4 depleted mice was significantly larger than tumors in CD8 depleted

mice at days 12, 15, 19 and 22, but significantly smaller than tumor size in double depleted mice

at every time point after day 9(Figure 13b and c).

80

These results suggest that both CD4+ and CD8+ T cells are required to fully protect against

rechallenge. CD4+ T cells are critically required for an effective memory response as 5/6

depleted animals failed to reject a tumor when rechallenged. However, tumor growth was

significantly delayed compared to double depleted mice suggesting CD8 lymphocytes can

protect the animal but need intact CD4 function to be fully protective. All CD8 depleted mice

completely rejected the tumors, suggesting that CD4 T cells are sufficient to mount an effective

response against the tumor, probably by recruiting other immune cells than CD8 T cells.

81

Figure 13. CD4+ and CD8+ T cells role during rechallenge. A) Mice that had previously

rejected a tumor after rechallenge were depleted of CD4+ T cells, CD8+ T cells or both and

rechallenged one more time. B) Tumor size was significantly larger in double depleted mice

compared to CD4 or CD8 single depleted mice. Tumor size was also significantly larger in CD4

depleted mice compared to CD8 depleted mice but smaller than double depleted mice. C) AE-

17-OVA tumor growth of double depleted mice is faster compared to CD4 or CD8 single

depleted mice. Values shown are the mean tumor area in mm2 of 6 mice per time point in the

CD4 group and 7 mice in the other groups and are expressed as mean ± SEM. ** < 0.05,

compared to CD8, §<0.005, compared to CD4.

A

B C

82

4.4 CTLA-4 blockade improves the beneficial effect of LRT on tumors

In this experiment, the goal was to determine the role of anti-CTLA-4 combined with LRT in

improving AE17-OVA tumor growth control. CTLA-4 is a negative regulator of T cell activation

and blockade of its function induces effective antitumor immunity. Therefore, the combination of

LRT and anti-CTLA4 may show a synergistic effect. Tumor bearing C57BL/6 mice were

randomized and received 1) No treatment, 2) anti-CTLA4, 3) LRT, 4) LRT and anti-CTLA-4.

Each group consisted of 4 mice. Anti-CTLA-4 and LRT started on day 9 in all groups, and the

anti-CTLA-4 dose was repeated every 3 days for a total of 3 doses.

The group treated only with anti-CTLA4 was not significantly different than the untreated group.

Tumor size of mice treated with anti-CTLA-4 was significantly larger at day 16 and 18

compared to LRT, and at day 16, 18 and 21 compared to the combination group. Mice treated

with the combination group had significantly smaller tumors at day 18 and 21 compared to LRT

only. In a similar experiment where 5 mice were treated with LRT and anti CTLA-4 antibody we

observed consistent results and with longer follow up 2 out of 5 mice completely rejected the

tumor (results not shown) (Figure 14) .

Overall, blockade of CTLA-4 during radiation treatment showed a synergistic beneficial effect

on tumor growth. However, blockade of CTLA-4 with no LRT did not significantly affect tumor

growth.

83

Figure 14. Combination therapy with LRT and CTLA-4 shows a synergistic effect on

tumor growth. A) Schematic of the experiment. B) Tumor growth significantly slows down in

the combination group compared to the antiCTLA-4 group and to the LRT group. C)

Combination therapy is significantly better at controlling tumor size at day 18 and 21. Values

shown are the mean tumor area in mm2 of 4 mice per time point in all groups and are expressed

as mean ± SEM. ** < 0.05, compared to LRT, §<0.005, compared to anti-CTLA-4.

84

5 Discussion

Radiation therapy is an important treatment modality for mesothelioma and cancer in general.

Until recently, the beneficial effect was based on the ability of radiation to eradicate cancer cells

by means of its cytotoxic effect (Demaria, Bhardwaj, et al. 2005). However, the effects of

radiation may extend beyond the elimination of radiosensitive tumor cells. Although radiation

therapy is a localized therapy, it can have a major impact on the immune system and have a

systemic effect. Considerable evidence indicates that radiation therapy has the potential to

enhance tumor immunogenicity by promoting cross priming and eliciting antitumor T-cell

responses (Demaria et al. 2004). The generation of inflammation and modification of the tumor

microenvironment after RT results in an immunogenic milieu which could provide a unique

opportunity to combine radiation and immunotherapy to obtain a synergistic effect on the tumor

killing. Furthermore, recent studies have shown that local radiation can eradicate the targeted

tumor, but also distant metastasis away from the radiation field through specific activation of the

immune system against the targeted tumor cells, this is called the abscopal effect (B. Park, Yee,

and Lee 2014). The abscopal mechanism remains unexplained, although a variety of underlying

biologic events can be hypothesized, including a possible role for the immune system.

The aims of this project were to evaluate the effect of Local Radiation Therapy (LRT) combined

with surgery in a mouse model of mesothelioma and to investigate the involvement of the

immune system. As mentioned before, radiation may have a systemic impact on the immune

system by activating immune cells, and, by surgically removing the tumor, the

immunosuppressive factors produced by the tumor tissue may add to the immunogenic effect of

85

LRT. Lastly, the addition of immunotherapeutic molecules may shift the balance toward a strong

immune response against the tumor. Thus, in this study 1) a mouse model of LRT followed by

surgery was developed, 2) CD8+ T cells were analyzed within the tumor after LRT, and 3) the

effect of immunotherapy together with LRT was examined in this model.

5.1 Development of a mouse model of MPM treated with LRT

followed by surgery

The first part of the project involved the development of the mouse model. A previous mouse

model used by our group was examined initially. This model consisted of BALB/c mice

inoculated subcutaneously in the right flank or leg with 2x106 AB12 cells. The number of

inoculated cells was lowered to 1x106 AB12 cells to slow tumor growth. This change resulted in

slower tumor growth and implantation success was 100% (data not shown). The next step was to

evaluate if the tumor was sensitive to radiation and to find the best radiation dose and regimen.

Multiple studies have shown evidence that LRT at typical doses consistently elicits some

activation of the innate and adaptive immune system (McBride et al. 2004; A. a Lugade et al.

2005; Keisari et al. 2014; Demaria et al. 2004). On the other hand, the proportion of cells

undergoing immunogenic cell death and the recruitment of inhibitory cells versus DCs is

variable depending on the dose and fractionation (Demaria and Formenti 2012; Klug et al. 2013).

Also, some authors speculate that antigens released after radiation-induced-cell death need to be

pulsed to overcome a critical threshold to achieve antitumor immune responses (Frey et al.

2014). Several recent studies have shown that hypofractionated doses are more effective at

activating the immune system and eliciting an abscopal effect with in-vivo models and in clinical

86

reports compared to standard radiation (Dewan et al. 2009; Camphausen et al. 2003; Golden et

al. 2013; Postow et al. 2012; Victor et al. 2015; Filatenkov et al. 2015). Similar benefits have

been demonstrated by others with single ablative dose regimens (Reits et al. 2006; Demaria,

Kawashima, et al. 2005; Shiraishi et al. 2008; Matsumura et al. 2008). Overall multiple

preclinical and clinical publications support the immunogenic effect of radiation but there is no

consensus about the best radiation regimen to be used for the best antitumor immune response.

Finding the optimal regimen may affect the ability of radiation to make an in-situ vaccine out of

a tumor.

All doses evaluated in our model provided good tumor control but the highest doses resulted in

side effects as demonstrated by weight loss and animal distress. Although the literature is not

decisive, we favor in our group the use of hypofractionated radiation as the better approach for

activating the immune system since it is clinically relevant. Furthermore, the 15Gy dose

fractionated over 3 days mimics more closely the SMART protocol and is consistent with other

projects in our group.

Lastly, the combination therapy with LRT and Surgery was evaluated in the mouse model. A

significant increase in tumor growth rate and a detrimental effect on survival was observed in the

group treated with blunt surgery alone compared to untreated and the LRT groups. The increase

in tumor growth rate can be explained by the cytokines and growth factors released at the site of

the excised tumor (Kal, Struikmans, and Barten-van Rijbroek 2008; Tsuchiya et al. 2003).

Among the many molecules present at the surgical wound, transforming growth factor beta

(TGF-β)(Hofer et al. 1998), epidermal growth factor (EGF) (Tagliabue et al. 2003), vascular

endothelial growth factor (VEGF) (Hormbrey et al. 2003) and basic fibroblast growth factor,

importantly promote tumor growth in addition to promoting healing on the surgical wound.

87

These factors promote neoangiogenesis, wound healing and cancer cell proliferation (Demicheli

et al. 2008). Furthermore, dysfunction of the immune system may play a role in the progression

of tumors after surgical stress (Menges et al. 2012). The postoperative inflammatory state

induces inhibition of cell-mediated immunity (Cardinale et al.), the release of glucocorticoids, a

reduced rate of T cell proliferation, and lymphocytopenia (Dhabhar et al. 1996; Ogawa et al.

2000), and activation of the sympathetic nervous system results in catecholamine mediated

inhibition of NKT cells (I J Elenkov et al. 2000; Rosenne et al. 2014). Thus, cytokines and

growth factors promoting tumor growth and factors depressing the immune response may

explain the increased rate of tumor growth after blunt surgery.

The LRT group had a significant decrease in tumor growth compared to blunt surgery and

untreated groups. Tumor growth slowed down significantly, but it only lasted for 7 to 10 days

before tumor growth accelerated once more. As discussed previously there is growing evidence

that support the immunogenic effect of radiation. However, the demand faced by the immune

system by a rapidly growing tumor is challenging. Clinically detectable tumors double in size in

a period of time measured in weeks or months, but tumors treated with radiation can progress

faster with a doubling time in the order of days. It has been reported that accelerated growth of

some tumor cells surviving radiation proceeds at a rate 20 times faster than before treatment

(Withers 1993; Yom 2015; Abe et al. 1991). As a result, without debulking and inhibition of

tumor cell repopulation, the immune system may not be capable of eliminating or controlling the

tumor.

In our model, in the combined therapy group with LRT and surgery, tumor growth was

significantly delayed compared to the untreated and blunt surgery group. In the combination

group, tumor growth was delayed before and after blunt surgery and one of five mice was cured.

88

Hence, the combination therapy abrogates the negative effect observed in the group treated with

surgery alone and may have the advantage of removing the immunosuppressive tumor bulk, thus

promoting the benefit of LRT activating the immune system. Adding immunotherapy to further

activate the immune response against the rapidly proliferating tumor cells might be beneficial to

balance the immune response towards a cytotoxic immune response.

5.2 T cell tumor infiltration

When tumors were analyzed for CD8 T cell infiltration, we observed in the radiated tumor 2

days after treatment an important decrease in infiltrating T cells. This was most likely secondary

to the cytotoxic effect of radiotherapy as T cells are extremely sensitive to radiation (Manda et

al. 2012; Heylmann et al. 2014). However, although radiation may initially kill lymphocytes, it

does not have systemic cytotoxic effects and lymphocytes from the systemic pool re-infiltrate the

tumor not long after. As we observed in our experiment, at day 7, treated tumors had been

repopulated and at day 12 we observed a dramatic increase in infiltrating T cells. This contrasts

with the progressive decrease in infiltrating lymphocytes in the entire tissue section of untreated

tumor. We further demonstrated that this rise in tumor infiltrating lymphocytes after LRT was

antigen specific as we found an increase in OVA-tetramer CD8+ T cells in the radiated tumor.

Lymphocyte trafficking and infiltration into solid tumors secondary to radiation has been

examined in different murine models. Lugade et. al., treated B16-OVA tumors and this led to

increased priming of tumor specific T cells in the draining lymph node and increased infiltration

of CD8+ T cells in the tumor (A. a Lugade et al. 2005). Also, OT1 CD8+ T cells specific for

OVA that were activated ex-vivo and adoptively transferred were directed to the radiated tumor.

89

In a different model, Takeshima et. al. reported increased number of tumor specific T cells

infiltrating the tumor and draining lymph nodes following radiation, but a major reduction

despite the radiation in mice whose lymph nodes were surgically ablated or genetically defective

(Takeshima et al. 2010). Thus, radiation of a tumor promotes tumor infiltration by lymphocytes.

Radiation creates a pro inflammatory environment where danger signals and cytokines are

abundant creating a concentration gradient that directs the migration of lymphocytes towards the

tumor (A. a Lugade et al. 2005). One of the cytokines that play an important role is IFN-γ (B.

Park, Yee, and Lee 2014). Mice deficient in IFN-γ are unable to upregulate expression of

VCAM-1 and MHC class 1 in the tumor vasculature after radiotherapy, important for T cell

infiltration and tumor cell target recognition (A. A. Lugade et al. 2008). ICAM-1 is also

upregulated after radiation in human colon cancer and gastric adenocarcinoma cells (Chiriva-

Internati et al.). ICAM-1 facilitates lymphocyte infiltration. Secretion of chemokines such as

CCL2 (Draghiciu et al. 2014) and CXCL16, which bind CXCR6 on Th1 and CD8 T cells, are

increased after radiation in mouse and human breast cancer cells, and this leads to increased

migration of CD8 T cells to the tumor (Matsumura et al. 2008). This evidence suggest that

radiation of a tumor may direct lymphocytes to the tumor by upregulation of adhesion molecules

and an increase in the secretion of chemokines.

Finally, decreasing number of infiltrating T cells in the untreated tumor was observed over time.

Initially the tumor was infiltrated by a similar number of T cells probably by the initial immune

response after tumor cell injection. However, the immunosuppressive environment of the tumor

may have led to anergy and apoptosis of tumor infiltrating T cells in the untreated tumors. The

immunosuppressive tumor environment involves infiltrating T regulatory cells, M2

macrophages, PD-L1 and TGF-β expression. Also the downregulation of MHC and loss of tumor

90

associated antigen expression, allow the tumor to evade specific immune recognition. Radiation

may also reverse this immunosuppressive state by the release of proinflammatory molecules.

5.2.1 Upregulation of the activation marker 4-1BB and decrease in the

exhaustion marker PD-1

Tumor specific infiltrating lymphocytes were further analyzed on days 3 and 7 after local

radiation therapy and compared with untreated tumors as controls. At day 3 there was a

significant upregulation of the activation marker 4-1BB and downregulation of PD-1. At day 7,

4-1BB returned to similar levels as the untreated tumor but persistent lower expression of PD-1

was observed. 4-1BB is mainly present in activated T cells but not resting T cells and has

important effects on T cell proliferation and function. Signaling through 4-1BB in lymphocytes

can stimulate in a CD28 independent way, production of IL-2, a critical step for activation of T

cells but also for prevention of an anergic state (Saoulli et al. 1998). 4-1BB prevents apoptosis

and increase survival of tumor infiltration lymphocytes via upregulation of the antiapoptotic

pathways involving expression of Bcl-x and Bfl-1 (H.-W. Lee et al. 2002). In addition, 4-1BB

also enhances cytotoxic T lymphocyte cytolytic activity (Hernandez-Chacon et al. 2011). PD-1

on the other hand identifies T cells with an exhausted phenotype that cannot maintain appropriate

cytokine production (Fourcade et al. 2010). PD-1 in physiological conditions plays a role in

limiting damage to normal tissue during inflammatory response (Topalian, Drake, and Pardoll

2012). One of its ligands, PD-L1 is common in tumors including mesothelioma and has been

shown to correlate with poor prognosis (Cedrés et al. 2015). Furthermore, intratumoral

infiltration of PD-1 positive T cells was also correlated with progression of malignant tumors in

91

humans (Richendollar et al. 2011; Thompson et al. 2007). Also, PD-1 upregulation on CD8 T

cells is associated with T cell dysfunction, low proliferation and low activation (Keir et al. 2008).

In our experiment, the high expression of PD-1 and low expression of 4-1BB suggest

lymphocytes in untreated tumors may have been initially recognized tumor antigens but were no

longer in an activated state. The decreased PD-1 expression in tumor specific infiltrating

lymphocytes at days 3 and 9, together with the initial decrease in CD8+ T cells 2 days after

radiation, suggest radiation eliminated exhausted lymphocytes and new CD8+ T cells

repopulated the tumor. Again, the pro-inflammatory environment after irradiation may have been

responsible for the increased expression of the activation marker 4-1BB. Overall, radiation

therapy led to increased tumor-specific lymphocyte infiltration and these lymphocytes expressed

higher activation markers and downregulated the exhaustion marker PD-1. These lymphocytes

may have been responsible for at least part of the beneficial effect of radiation.

5.2.2 Depletion of lymphocytes partially abrogated the effect of radiation

Double depletion of CD4+ and CD8+ T cells during hypofractionated radiation significantly

abolished the effect of radiation. Depletion of CD8+ T cells partially abolished the therapeutic

effect of radiation as shown by the trend towards increased tumor growth rate compared to

radiation alone. Depletion of CD4+ T caused a trend towards the opposite direction, insinuating

an improved effect of radiation. These findings suggest both CD4+ and CD8+ T cells play a

major role in the response to radiation. While the direct cytotoxic effect of radiation was

preserved in all depleted groups, based on our results, CD4+ and CD8+ T cells together appear

to account for at least a third of the response to radiation. While depletion of CD8+ T cell did not

significantly reduced the effect of radiation, we noted a strong trend towards faster tumor growth

92

compared to radiation only. Also, the difference between the CD4 depleted and CD8 depleted

group was significant, suggesting CD8+ T cells might be responsible for large part of the lost

therapeutic effect in the double depleted mice. Regulatory T cells (CD4+CD25+FoxP3+) were

also depleted in the CD4 depleted group and that might explain the trend towards better tumor

control after radiation as reported by others including our group (Licun Wu et al. 2011; Son et al.

2015). In this CD4 depleted group, CD8+ T cells might have been activated by DCs exposed to

danger signals in the strong inflammatory tumor environment after radiation without the help of

CD4 T cells. Strengthening this idea, other groups have shown that this activation of CD8 T cells

without help of CD4 T helper leads to impaired CD8 memory responses (Novy et al. 2007; Sun,

Williams, and Bevan 2004). Our observations are in line with the literature indicating CD8 T

cells are required for the therapeutic effects of ablative radiation (Y. Lee et al. 2009; Gupta et al.

2012).

5.3 Protective memory response after LRT and Surgery

LRT was shown to result in the induction of protective immune memory response against the

tumor as demonstrated in the rechallenge experiments 90 days after LRT followed by radical

surgery. The group treated with radical surgery and the group treated with LRT followed by

radical surgery after 24 hrs displayed an accelerated tumor growth rate compared to LRT

followed by radical surgery after 7 days. In addition we did not observe rejections in these two

groups. By contrast, 3 out 10 mice that were treated with hypofractionated radiation 7 days

before radical surgery rejected the tumor, and the other 7 mice had a significantly reduced tumor

93

growth rate compared to the other 2 groups. The cell line AE17-OVA was weakly immunogenic,

as demonstrated by the fact that radical surgery does not provide protective memory.

Overall LRT before surgery contributed to generating a partially protective immunological

memory, but only when surgery was performed after 7 days and not when it was performed after

24 hrs. This might be as 7 days are required to reach the peak of the adaptive T cell immune

response in mice (Busch et al. 1998). Others have shown that, 5 days after RT, there is an

increase in DC loaded with tumor peptides in the draining lymph node and in the tumor (Y. Lee

et al. 2009). Together, these findings suggest that radiation might activate DCs within the tumor,

and then promotes maturation and migration of DCs to the draining lymph node to present

antigens and activate T cells which then will migrate to the tumor. This is in agreement with our

CD8+ T cell infiltration experiment where we observed important repopulation of T cells within

the tumor at 7 and 12 days but not at 2 days.

5.3.1 Depletion and rechallenge

Mice previously protected against AE17-OVA failed to reject the tumor against rechallenge

when CD4+ and CD8+ T cells were depleted and when CD4+ alone was depleted. CD4+

depletion preserved some protection against the tumor as 1 out of 6 mice rejected the tumor and

the tumor growth rate curve was slower.

These results suggest that both T cell subsets are required for an optimal protection. However,

for memory response, CD4 T cells are the key players. As mentioned in the depletion experiment

during LRT, there are some situations where CD8 T cells can be activated by mature DCs

without the help of CD4 T helper cells during the primary response (Novy et al. 2007;

94

Andreasson et al. 2010). However, these CD8 T cells will not mount a strong memory response

if rechallenged. Not only are CD4 T cells important during the primary encounter to create an

adequate memory CD8 T cell response, but they are crucial during the memory response to

expand and activate the pool of memory CD8 T cells. Despite the fact that all CD8 depleted

animals completely rejected the tumor, CD8 T cells are important during the memory response,

but they need CD4 T cell help. Perhaps, in the CD8 depleted mice, thanks to redundancy in the

immune system and after the very efficient tumor vaccine created in situ by LRT, CD4 T cells

orchestrated the memory immune response and activated other effector cells such as NKT cells

and macrophages and that was enough to reject the tumor.

5.4 CTLA-4 blockade synergized with LRT

Finally, we observed improvement of the therapeutic effects of hypofractionated radiation when

the immune checkpoint CTLA-4 was blocked. We observed a significant and dramatic

synergistic effect in the group treated with the combination therapy but no effect in the group

treated with the antibody alone. The observation that in the absence of an active immune

response, the beneficial effect of anti-CTLA-4 is minimal has been reported before by our group

and others, suggesting that combination with other therapeutic modalities is important (Licun

Wu et al. 2015; Davila, Kennedy, and Celis 2003; Grosso and Jure-Kunkel 2013; Vanpouille-

Box et al. 2015). It is believed anti-CLTA-4 therapy is not effective as a single agent because of

the dominant immunosuppressive tumor environment. Radiation and its capacity to create

inflammation and immunogenic cell death might be an excellent therapeutic adjunct to CTLA-4

blockade. This idea together with the dramatic effect of CTLA-4 in combination with LRT in

95

preclinical models is recognized and has emerged as a promising strategy in the clinical setting.

Since 2011, ipilimumab (Bristol-Meyers Squibb) an anti-CTLA-4 drug was approved for patients

with metastatic or unresectable melanoma and there are some encouraging reports. In MPM,

Tremelimumab therapy (AstraZeneca) has been explored in patients with unresectable disease

and results were published recently (Calabrò et al. 2013). Furthermore, a large multicenter

randomized trial with Tremelimumab was initiated in patients with unresectable and

chemotherapy resistant mesothelioma. However, currently there are no trials in MPM using

CTLA-4 blockade in combination with LRT.

96

6 Conclusions

The goal of this project was to develop a mouse model to recreate the effects seen with SMART

treatment in the clinic and asses the immunogenic effect of local hypofractionated radiation in a

model of malignant mesothelioma. It was suggested in the SMART trial that the benefits

observed in patients might have been related to activation of the immune system. The purpose of

the model was to investigate this suggestion and to find ways of improving it.

First, tumor sensitivity to radiation together with a safe dose and radiation regimen was assessed.

Several recent studies have shown that hypofractionated doses might be more effective at

activating the immune system and eliciting an abscopal effect compared to standard fractionated

radiation. Single dose and fractionated regimens were compared in the first part of the project.

All doses provided good and similar tumor control but the highest doses resulted in unacceptable

side effects in the animals. To be consistent with other projects in our group and to mimic more

closely the SMART protocol, 15 Gy over 3 days was chosen as the dose and regimen for the rest

of the project.

Afterwards, a subcutaneous mouse model was developed where we combined local radiation

therapy and surgery and we obtained similar results as seen in the clinic. LRT caused significant

tumor growth deceleration for a brief period of time. When the tumor was surgically removed,

the beneficial effect of local radiation therapy was preserved for a period of time. This was in

contrast to mice treated with blunt surgery only, where a significant increase in tumor growth

rate and detrimental effect on survival was observed after surgical removal of the tumor. The

beneficial effect provided by local radiation therapy before surgical removal of a tumor might be

97

secondary to immune activation. Furthermore, combination therapy abrogated the negative effect

observed in the group treated with surgery alone and may have the advantage of removing the

immunosuppressive tumor bulk, thus promoting the benefit of LRT activating the immune

system. Alternatively, the tumorostatic effect of radiation may have prevented or delayed

successful implantation and proliferation of tumor cells after surgery.

Furthermore, we analyzed tumors to elucidate if there was evidence of an involvement of the

immune system. We found an increase in tumor infiltrating T cells in the radiated tumor after 7

days. Conversely, in untreated tumors there was a distinct lack of tumor infiltrating T cells, likely

secondary to the strong immunosuppressive environment of the tumor. The tumor environment

may lead to anergy and apoptosis of tumor infiltrating T cells in the untreated tumors. A large

number of the lymphocytes in the radiated tumor were specific against the tumor and exhibited

an activated profile whereas untreated tumors upregulated exhaustion markers on T cells. These

findings suggest a strong activity of the immune system in our model after hypofractionated

radiation.

Radiation creates a pro inflammatory environment with abundant danger signals and cytokines,

providing a concentration gradient that attracts lymphocytes into the tumor. Radiation also

causes upregulation of adhesion molecules present on the tumor blood vessels and increased

secretion of chemokines. Together these effects after radiation of a tumor directs lymphocytes to

the tumor and favor its activity against tumor cells.

Depletion experiments supported the role of the immune system on the therapeutic response of

radiation therapy and the memory protection provided by radiation and surgery. Animals treated

with hypofractionated radiation and surgery were partially protected to tumor rechallenge,

compared to mice treated only with surgery which were not protected. This protection against

98

tumor rechallenged was lost after depleting CD4+ and CD8+ lymphocytes on mice previously

protected.

Lastly, we observed an improvement of the therapeutic effect of hypofractionated radiation

when the immune checkpoint CTLA-4 was blocked. A dramatic synergistic effect was observed

in the group treated with radiation and CTLA-4 blockade while single therapy with anti CTLA-4

showed no difference when compared with untreated mice. This lack of effect of anti CTLA-4

therapy might be secondary to the dominant immunosuppressive tumor environment. However,

when tumors are treated with radiation the transient inflammatory milieu might be a trigger to

initiate an immune response against the tumors. This synergistic effect of radiation and blockade

of the immune checkpoint inhibitor anti-CTLA-4 supported the potential benefit of

immunomodulation of the immune system in combination with hypofractionated radiation.

Malignant Pleural Mesothelioma remains a major public health concern and at present only few

patients are candidate to the potentially curative surgery within a multimodality treatment

including chemotherapy and radiotherapy. Also, there is no second line treatment that

significantly prolongs survival of patients. Thus new alternatives are needed.

The goal of this project was to translate findings into clinical practice. In vivo date suggests that

radiation is effective in creating an inflammatory tumor environment and recruiting immune cells

into the tumor. Radiation therapy is emerging as an optimal partner for immunotherapy such as

anti CTLA-4 because of its ability to induce immune activation. Radiation immunogenicity and

combination with immunotherapy warrants further investigation and supports the potential

benefit of immunomodulating therapy in combination with hypofractionated radiation.

99

Future development of clinical trials directed at the combination of radiation, surgery and

immunotherapy will potentially open new avenues in the treatment of mesothelioma.

100

7 Limitations

Although the project started with the weakly immunogenic cell line AB12, most of the

experiments were performed with the cell line AE17-OVA. Although this cell line is not highly

immunogenic, as mice treated with radical surgery did not reject the tumor after rechallenge, the

artificially transfected OVA antigen does increase its immunogenicity. It may be worth,

repeating these experiments with the AB12 mouse model.

A limitation with animal models and cell lines is the difference in the tumor microenvironment

and subsequent immune editing as compared to spontaneous tumor. In spontaneous tumors, it

takes long periods of inflammation to develop a tumor whereas injection of cells takes a few

days. This may lead to important differences in the tumor environment and to the immune

response against the tumor.

Another limitation of our model is the subcutaneous location of the tumor. The connective tissue

and blood supply found in the subcutaneous space is certainly different than the pleural space.

This might affect the response to radiation and the effect on immune cell migration.

In regards to tumor specific T cells, significant differences were not found, most likely as a result

of the number of animals analyzed and the single time point used. It may be beneficial to

increase the number of animals and repeat the analysis on the same time points used for

immunofluorescent staining at 2, 7 and 12 days. Other difficulties arose while analyzing tumor

infiltrating lymphocytes as tumors can differ in size dramatically. Untreated tumors are up to 10

times larger than radiated tumors. While disrupting tumors into a single cell suspension,

101

lymphocytes might be artificially concentrated and the number overestimated in untreated

tumors.

Finally the cytotoxic activity of infiltrating lymphocytes was not investigated. The fact that there

is an increase number of T cells may not be relevant if they are unable to lyse tumor cells. Future

studies will need to look at the functional status of cytotoxic T cells and their capacity to secrete

relevant cytokines to achieve their role.

102

Future Directions

There is no consensus about the best radiation regimen to be used for the best antitumor immune

response. It would be interesting to compare the ability of single-dose, hypofractionated and

fractionated radiation and its ability to activate the immune response. Finding the optimal

regimen for combination therapy remains to be done.

An intrapleural model would be a more clinically relevant model of MPM. As it was discussed in

the limitations there are several weaknesses in our subcutaneous model. Injection of tumor cells

in the thoracic cavity under direct visualisation can be done through a small incision under

anesthesia. Tumor growth would be more difficult to monitor, but it is possible to perform CT or

PET scan to have an approximate evaluation of the tumor growth. For radiotherapy, the XRAD

225cx would allow us to provide hypofractionated hemithoracic radiation to a mouse intrapleural

tumor without compromising other vital organs. Finally, pneumonectomy could be performed

with a similar technique to what is done in lung transplant model by others in the Thoracic

Surgery Research Laboratories.

Also to resemble more closely to what is seen in human MPM, future studies could involve

mouse MPM developed from asbestos exposure. Spontaneously developed tumors may be less

immunogenic than cell lines due to slow selection of cancer cell clones that escape the immune

system, potentially better modelling clinical cases of MPM. Spontaneous development is

possible as asbestos tumor development in mice has been extensively demonstrated. However,

long incubation period and low number of mice that develop tumors after asbestos exposure still

poses a potential challenge.

103

To assess the systemic effect of immune activation after hypofractionated radiation a metastatic

cell line can be used. AB12 and AE17 are poorly metastatic in vivo and systemic protection

against the tumor cannot be assessed properly. To simulate metastasis in a different mouse model

in our group a tumor is injected in two sites in the same animal at the same time point. Only one

of the two sites is treated with radiation and the systemic effect is evaluated in the untreated

tumor. However, this model does not resemble closely the biology of spontaneous metastatic

tumors. To overcome this difficulty we could utilize a spontaneous metastatic cell line that have

been reported in other preclinical tumor models such as breast cancer. The limitation with such a

cell line is that there is no metastatic mesothelioma cell line reported in the literature.

Other alternative to support our findings on the beneficial effect of combination therapy with

radiation and surgery, could be a different animal model. Other authors have reported

intrapleural mesothelioma in rats. Similar results in different species could provide stronger

evidence for the beneficial immunogenic effect of radiation therapy.

Lastly, as mentioned previously beside mesothelin and WT-1 there are only a few known

specific malignant pleura mesothelioma associated tumor antigens. Finding other mesothelioma

associated tumor antigens might be useful to implement immunotherapeutic approaches in MPM

patients.

In regards to infiltrating lymphocytes into the tumors, we have not analyzed the functional status

of T cells. CD8 T cells may be present in the tumor in an anergic state and unable to lyse tumor

cells. An important future experiment involves the analysis of T cells infiltrating the tumor. Flow

cytometry analysis and PCR analysis to investigate the capacity of T cells to secrete cytokines

such as IFN γ and to produce granzyme B and perforin to lyse tumor cells is the next step in this

project.

104

The SMART approach in patients involved radiation of mesothelioma before extrapleural

pneumonectomy. Previously, adjuvant radiation was done after extrapleural pneumonectomy.

The rationale on SMART beneficial effect was that the irradiated tumors released danger signals

and tumor associated antigens before tumor removal causing activation and priming of the

immune system. In this project a mouse model was stablished and we demonstrated longer

survival in mice treated with the SMART approach and increased infiltration of lymphocytes

within the tumor. To further understand if the increase in survival in patients treated with

SMART is immune mediated, tumors infiltrating lymphocytes could be measured and analyzed

for activation and exhaustion markers after the completion of treatment. Tumor of SMART

treated patients could be compared with patients treated with extrapleural pneumonectomy

without radiotherapy.

Finally, the following and ultimate goal of this project would be to translate this findings into the

clinic. A clinical trial comparing patients treated with SMART vs SMART combined with

immunotherapy could be considered. Similar trials involving immune checkpoint inhibitors are

undergoing in other tumors in patients. However, to the best or our knowledge there are no

undergoing clinical trial involving hypofractionated radiation and immune checkpoint inhibitors

in MPM patients.

105

8 References

Abe, Y, M Urano, L A Kenton, J Kahn, and C G Willet. 1991. “The Accelerated Repopulation of

a Murine Fibrosarcoma, FSA-II, during the Fractionated Irradiation and the Linear-

Quadratic Model.” International Journal of Radiation Oncology, Biology, Physics 21 (6):

1529–34. http://www.ncbi.nlm.nih.gov/pubmed/1938563.

Adams, V I, K K Unni, J R Muhm, J R Jett, D M Ilstrup, and P E Bernatz. 1986. “Diffuse

Malignant Mesothelioma of Pleura. Diagnosis and Survival in 92 Cases.” Cancer 58 (7):

1540–51. http://www.ncbi.nlm.nih.gov/pubmed/3742473.

Ahamad, Anesa, Craig W Stevens, W Roy Smythe, Ara A Vaporciyan, Ritsuko Komaki, Jason F

Kelly, Zhongxing Liao, George Starkschall, and Kenneth M Forster. 2003. “Intensity-

Modulated Radiation Therapy: A Novel Approach to the Management of Malignant Pleural

Mesothelioma.” International Journal of Radiation Oncology, Biology, Physics 55 (3):

768–75. http://www.ncbi.nlm.nih.gov/pubmed/12573764.

Albert, M L, S F Pearce, L M Francisco, B Sauter, P Roy, R L Silverstein, and N Bhardwaj.

1998. “Immature Dendritic Cells Phagocytose Apoptotic Cells via alphavbeta5 and CD36,

and Cross-Present Antigens to Cytotoxic T Lymphocytes.” The Journal of Experimental

Medicine 188 (7): 1359–68.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2212488&tool=pmcentrez&ren

dertype=abstract.

Alberts, Bruce, Alexander Johnson, Julian Lewis, Martin Raff, Keith Roberts, and Peter Walter.

2002. “Lymphocytes and the Cellular Basis of Adaptive Immunity.” Garland Science.

http://www.ncbi.nlm.nih.gov/books/NBK26921/.

Amin, A M, C Mason, and P Rowe. 2001. “Diffuse Malignant Mesothelioma of the Peritoneum

Following Abdominal Radiotherapy.” European Journal of Surgical Oncology : The

Journal of the European Society of Surgical Oncology and the British Association of

Surgical Oncology 27 (2): 214–15. doi:10.1053/ejso.2000.1024.

106

Andreasson, Kalle, Mathilda Eriksson, Karin Tegerstedt, Torbjörn Ramqvist, and Tina Dalianis.

2010. “CD4+ and CD8+ T Cells Can Act Separately in Tumour Rejection after

Immunization with Murine Pneumotropic Virus Chimeric Her2/neu Virus-like Particles.”

PLoS ONE 5 (7). doi:10.1371/journal.pone.0011580.

Anraku, Masaki, Kristopher S Cunningham, Zhihong Yun, Ming-Sound Tsao, Li Zhang, Shaf

Keshavjee, Michael R Johnston, and Marc de Perrot. 2008. “Impact of Tumor-Infiltrating T

Cells on Survival in Patients with Malignant Pleural Mesothelioma.” The Journal of

Thoracic and Cardiovascular Surgery 135 (4): 823–29. doi:10.1016/j.jtcvs.2007.10.026.

Antman, K, R Shemin, L Ryan, K Klegar, R Osteen, T Herman, G Lederman, and J Corson.

1988. “Malignant Mesothelioma: Prognostic Variables in a Registry of 180 Patients, the

Dana-Farber Cancer Institute and Brigham and Women’s Hospital Experience over Two

Decades, 1965-1985.” Journal of Clinical Oncology : Official Journal of the American

Society of Clinical Oncology 6 (1): 147–53. http://www.ncbi.nlm.nih.gov/pubmed/3335886.

Arenberg, D A, S L Kunkel, P J Polverini, S B Morris, M D Burdick, M C Glass, D T Taub, M D

Iannettoni, R I Whyte, and R M Strieter. 1996. “Interferon-Gamma-Inducible Protein 10

(IP-10) Is an Angiostatic Factor That Inhibits Human Non-Small Cell Lung Cancer

(NSCLC) Tumorigenesis and Spontaneous Metastases.” The Journal of Experimental

Medicine 184 (3): 981–92.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2192788&tool=pmcentrez&ren

dertype=abstract.

Ascierto, Paolo A. 2013. “Ipilimumab in the Treatment of Metastatic Melanoma: A Summary of

Recent Studies.” Tumori 99 (6): 302e – 5e. doi:10.1700/1390.15474.

Astoul, P, D Picat-Joossen, J R Viallat, and C Boutin. 1998. “Intrapleural Administration of

Interleukin-2 for the Treatment of Patients with Malignant Pleural Mesothelioma: A Phase

II Study.” Cancer 83 (10): 2099–2104. http://www.ncbi.nlm.nih.gov/pubmed/9827714.

Bachmayr-Heyda, Anna, Stefanie Aust, Georg Heinze, Stephan Polterauer, Christoph Grimm,

Elena Ioana Braicu, Jalid Sehouli, et al. 2013. “Prognostic Impact of Tumor Infiltrating

107

CD8+ T Cells in Association with Cell Proliferation in Ovarian Cancer Patients--a Study of

the OVCAD Consortium.” BMC Cancer 13 (January): 422. doi:10.1186/1471-2407-13-422.

Baldini, Elizabeth H. 2004. “External Beam Radiation Therapy for the Treatment of Pleural

Mesothelioma.” Thoracic Surgery Clinics 14 (4): 543–48. doi:10.1016/S1547-

4127(04)00108-2.

Barcellos-Hoff, M H, R Derynck, M L Tsang, and J A Weatherbee. 1994. “Transforming Growth

Factor-Beta Activation in Irradiated Murine Mammary Gland.” The Journal of Clinical

Investigation 93 (2): 892–99. doi:10.1172/JCI117045.

Batista, Luís F Z, Wynand P Roos, Markus Christmann, Carlos F M Menck, and Bernd Kaina.

2007. “Differential Sensitivity of Malignant Glioma Cells to Methylating and

Chloroethylating Anticancer Drugs: p53 Determines the Switch by Regulating Xpc, ddb2,

and DNA Double-Strand Breaks.” Cancer Research 67 (24): 11886–95. doi:10.1158/0008-

5472.CAN-07-2964.

Beatty, G L, and Y Paterson. 2000. “IFN-Gamma Can Promote Tumor Evasion of the Immune

System in Vivo by down-Regulating Cellular Levels of an Endogenous Tumor Antigen.”

Journal of Immunology (Baltimore, Md. : 1950) 165 (10): 5502–8.

http://www.ncbi.nlm.nih.gov/pubmed/11067903.

Beatty, Gregory L, Andrew R Haas, Marcela V Maus, Drew A Torigian, Michael C Soulen,

Gabriela Plesa, Anne Chew, et al. 2014. “Mesothelin-Specific Chimeric Antigen Receptor

mRNA-Engineered T Cells Induce Anti-Tumor Activity in Solid Malignancies.” Cancer

Immunology Research 2 (2): 112–20. doi:10.1158/2326-6066.CIR-13-0170.

Beilin, Benzion, Yehuda Shavit, Evelyn Trabekin, Boris Mordashev, Eduard Mayburd,

Alexander Zeidel, and Hanna Bessler. 2003. “The Effects of Postoperative Pain

Management on Immune Response to Surgery.” Anesthesia and Analgesia 97 (3): 822–27.

http://www.ncbi.nlm.nih.gov/pubmed/12933409.

Ben-Eliyahu, S, G G Page, R Yirmiya, and G Shakhar. 1999. “Evidence That Stress and Surgical

Interventions Promote Tumor Development by Suppressing Natural Killer Cell Activity.”

108

International Journal of Cancer. Journal International Du Cancer 80 (6): 880–88.

http://www.ncbi.nlm.nih.gov/pubmed/10074922.

Ben-Eliyahu, Shamgar, Gayle G Page, and Steven J Schleifer. 2007. “Stress, NK Cells, and

Cancer: Still a Promissory Note.” Brain, Behavior, and Immunity 21 (7): 881–87.

doi:10.1016/j.bbi.2007.06.008.

Bennett, M, K Macdonald, S W Chan, J P Luzio, R Simari, and P Weissberg. 1998. “Cell

Surface Trafficking of Fas: A Rapid Mechanism of p53-Mediated Apoptosis.” Science (New

York, N.Y.) 282 (5387): 290–93. http://www.ncbi.nlm.nih.gov/pubmed/9765154.

Beseth, Bryce D, Robert B Cameron, Pamela Leland, Liang You, Frederick Varricchio, Robert J

Kreitman, Richard A Maki, David M Jablons, Syed R Husain, and Raj K Puri. 2004.

“Interleukin-4 Receptor Cytotoxin as Therapy for Human Malignant Pleural Mesothelioma

Xenografts.” The Annals of Thoracic Surgery 78 (2): 436–43; discussion 436–43.

doi:10.1016/j.athoracsur.2004.03.010.

Bezu, Lucillia, Ligia C Gomes-de-Silva, Heleen Dewitte, Karine Breckpot, Jitka Fucikova,

Radek Spisek, Lorenzo Galluzzi, Oliver Kepp, and Guido Kroemer. 2015. “Combinatorial

Strategies for the Induction of Immunogenic Cell Death.” Frontiers in Immunology 6

(January): 187. doi:10.3389/fimmu.2015.00187.

Bocchetta, M, and Michele Carbone. 2005. “SV40-Mediated Oncogenesis.” In Malignant

Mesothelioma: Pathogenesis, Diagnosis, and Translational Therapies, edited by H Pass,

Nicholas J. Vogelzang, and Michele Carbone, 1st ed., 34–59. New York, NY: Springer.

Bocchetta, M, I Di Resta, A Powers, R Fresco, A Tosolini, J R Testa, H I Pass, P Rizzo, and M

Carbone. 2000. “Human Mesothelial Cells Are Unusually Susceptible to Simian Virus 40-

Mediated Transformation and Asbestos Cocarcinogenicity.” Proceedings of the National

Academy of Sciences of the United States of America 97 (18): 10214–19.

doi:10.1073/pnas.170207097.

Boehm, U, T Klamp, M Groot, and J C Howard. 1997. “Cellular Responses to Interferon-

Gamma.” Annual Review of Immunology 15 (January): 749–95.

doi:10.1146/annurev.immunol.15.1.749.

109

Botha, A J, F A Moore, E E Moore, A Sauaia, A Banerjee, and V M Peterson. 1995. “Early

Neutrophil Sequestration after Injury: A Pathogenic Mechanism for Multiple Organ

Failure.” The Journal of Trauma 39 (3): 411–17.

http://www.ncbi.nlm.nih.gov/pubmed/7473901.

Brahmer, Julie R, and Drew M Pardoll. 2013. “Immune Checkpoint Inhibitors: Making

Immunotherapy a Reality for the Treatment of Lung Cancer.” Cancer Immunology

Research 1 (2): 85–91. doi:10.1158/2326-6066.CIR-13-0078.

Brenner, J, P P Sordillo, G B Magill, and R B Golbey. 1982. “Malignant Mesothelioma of the

Pleura: Review of 123 Patients.” Cancer 49 (11): 2431–35.

http://www.ncbi.nlm.nih.gov/pubmed/7074556.

Bryk, Jodie A, Petar J Popovic, Mazen S Zenati, Veronica Munera, John P Pribis, and Juan B

Ochoa. 2010. “Nature of Myeloid Cells Expressing Arginase 1 in Peripheral Blood after

Trauma.” The Journal of Trauma 68 (4): 843–52. doi:10.1097/TA.0b013e3181b026e4.

Burnet. 1970. “The Concept of Immunological Surveillance.” Progress in Experimental Tumor

Research 13 (January): 1–27. http://www.ncbi.nlm.nih.gov/pubmed/4921480.

Burnet, M. 1957. “Cancer: A Biological Approach. III. Viruses Associated with Neoplastic

Conditions. IV. Practical Applications.” British Medical Journal 1 (5023): 841–47.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1973618&tool=pmcentrez&ren

dertype=abstract.

Burnette, Byron, and Ralph R. Weichselbaum. 2013. “Radiation as an Immune Modulator.”

Seminars in Radiation Oncology 23 (4). Elsevier: 273–80.

doi:10.1016/j.semradonc.2013.05.009.

Busch, D H, I M Pilip, S Vijh, and E G Pamer. 1998. “Coordinate Regulation of Complex T Cell

Populations Responding to Bacterial Infection.” Immunity 8 (3): 353–62.

http://www.ncbi.nlm.nih.gov/pubmed/9529152.

Buschke, F. 1958. “Progress in Radiation Therapy: Introduction.” Progress in Radiation Therapy

1 (January): 1–12. http://www.ncbi.nlm.nih.gov/pubmed/13601954.

110

Calabrò, Luana, Aldo Morra, Ester Fonsatti, Ornella Cutaia, Giovanni Amato, Diana Giannarelli,

Anna Maria Di Giacomo, et al. 2013. “Tremelimumab for Patients with Chemotherapy-

Resistant Advanced Malignant Mesothelioma: An Open-Label, Single-Arm, Phase 2 Trial.”

The Lancet Oncology 14 (11): 1104–11. doi:10.1016/S1470-2045(13)70381-4.

Caldenhoven, E, P Coffer, J Yuan, A Van de Stolpe, F Horn, W Kruijer, and P T Van der Saag.

1994. “Stimulation of the Human Intercellular Adhesion Molecule-1 Promoter by

Interleukin-6 and Interferon-Gamma Involves Binding of Distinct Factors to a Palindromic

Response Element.” The Journal of Biological Chemistry 269 (33): 21146–54.

http://www.ncbi.nlm.nih.gov/pubmed/7914891.

Camphausen, Kevin, Marsha A Moses, Cynthia Ménard, Mary Sproull, Wolf-Dietrich Beecken,

Judah Folkman, and Michael S O’Reilly. 2003. “Radiation Abscopal Antitumor Effect Is

Mediated through p53.” Cancer Research 63 (8): 1990–93.

http://www.ncbi.nlm.nih.gov/pubmed/12702593.

Canman, C E, D S Lim, K A Cimprich, Y Taya, K Tamai, K Sakaguchi, E Appella, M B Kastan,

and J D Siliciano. 1998. “Activation of the ATM Kinase by Ionizing Radiation and

Phosphorylation of p53.” Science (New York, N.Y.) 281 (5383): 1677–79.

http://www.ncbi.nlm.nih.gov/pubmed/9733515.

Carbone, M, H I Pass, L Miele, and M Bocchetta. 2003. “New Developments about the

Association of SV40 with Human Mesothelioma.” Oncogene 22 (33): 5173–80.

doi:10.1038/sj.onc.1206552.

Carbone, M, H I Pass, P Rizzo, M Marinetti, M Di Muzio, D J Mew, A S Levine, and A

Procopio. 1994. “Simian Virus 40-like DNA Sequences in Human Pleural Mesothelioma.”

Oncogene 9 (6): 1781–90. http://www.ncbi.nlm.nih.gov/pubmed/8183577.

Carbone, Michele, Salih Emri, A Umran Dogan, Ian Steele, Murat Tuncer, Harvey I Pass, and Y

Izzettin Baris. 2007. “A Mesothelioma Epidemic in Cappadocia: Scientific Developments

and Unexpected Social Outcomes.” Nature Reviews. Cancer 7 (2): 147–54.

doi:10.1038/nrc2068.

111

Carbone, Michele, Robert A Kratzke, and Joseph R Testa. 2002. “The Pathogenesis of

Mesothelioma.” Seminars in Oncology 29 (1): 2–17.

http://www.ncbi.nlm.nih.gov/pubmed/11836664.

Carbone, Michele, Haining Yang, Harvey I Pass, Thomas Krausz, Joseph R Testa, and Giovanni

Gaudino. 2013. “BAP1 and Cancer.” Nature Reviews. Cancer 13 (3): 153–59.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3792854&tool=pmcentrez&ren

dertype=abstract.

Cardinale, F, I Chinellato, S Caimmi, D G Peroni, F Franceschini, M Miraglia Del Giudice, and

R Bernardini. “Perioperative Period: Immunological Modifications.” International Journal

of Immunopathology and Pharmacology 24 (3 Suppl): S3–12.

http://www.ncbi.nlm.nih.gov/pubmed/22014920.

Carlos, T M. 2001. “Leukocyte Recruitment at Sites of Tumor: Dissonant Orchestration.”

Journal of Leukocyte Biology 70 (2): 171–84.

http://www.ncbi.nlm.nih.gov/pubmed/11493608.

Carpenito, Carmine, Michael C Milone, Raffit Hassan, Jacqueline C Simonet, Mehdi Lakhal,

Megan M Suhoski, Angel Varela-Rohena, et al. 2009. “Control of Large, Established

Tumor Xenografts with Genetically Retargeted Human T Cells Containing CD28 and

CD137 Domains.” Proceedings of the National Academy of Sciences of the United States of

America 106 (9): 3360–65. doi:10.1073/pnas.0813101106.

Castagneto, Bruno, Silvia Zai, Diego Dongiovanni, Alberto Muzio, Sergio Bretti, Gianmauro

Numico, Mario Botta, and G Sinaccio. 2005. “Cisplatin and Gemcitabine in Malignant

Pleural Mesothelioma: A Phase II Study.” American Journal of Clinical Oncology 28 (3):

223–26. http://www.ncbi.nlm.nih.gov/pubmed/15923792.

Cedrés, Susana, Santiago Ponce-Aix, Jon Zugazagoitia, Irene Sansano, Ana Enguita, Alejandro

Navarro-Mendivil, Alex Martinez-Marti, Pablo Martinez, and Enriqueta Felip. 2015.

“Analysis of Expression of Programmed Cell Death 1 Ligand 1 (PD-L1) in Malignant

Pleural Mesothelioma (MPM).” PloS One 10 (3): e0121071.

doi:10.1371/journal.pone.0121071.

112

Ceresoli, G L, P A Zucali, M Mencoboni, M Botta, F Grossi, D Cortinovis, N Zilembo, et al.

2013. “Phase II Study of Pemetrexed and Carboplatin plus Bevacizumab as First-Line

Therapy in Malignant Pleural Mesothelioma.” British Journal of Cancer 109 (3): 552–58.

doi:10.1038/bjc.2013.368.

Chang, K, L H Pai, J K Batra, I Pastan, and M C Willingham. 1992. “Characterization of the

Antigen (CAK1) Recognized by Monoclonal Antibody K1 Present on Ovarian Cancers and

Normal Mesothelium.” Cancer Research 52 (1): 181–86.

http://www.ncbi.nlm.nih.gov/pubmed/1727378.

Chen, Benjamin P C, Naoya Uematsu, Junya Kobayashi, Yaniv Lerenthal, Andrea Krempler,

Hirohiko Yajima, Markus Löbrich, Yosef Shiloh, and David J Chen. 2007. “Ataxia

Telangiectasia Mutated (ATM) Is Essential for DNA-PKcs Phosphorylations at the Thr-

2609 Cluster upon DNA Double Strand Break.” The Journal of Biological Chemistry 282

(9): 6582–87. doi:10.1074/jbc.M611605200.

Chen, Daniel S., and Ira Mellman. 2013. “Oncology Meets Immunology: The Cancer-Immunity

Cycle.” Immunity 39 (1): 1–10. doi:10.1016/j.immuni.2013.07.012.

Chen, Guoqian, Mary F Ward, Andrew E Sama, and Haichao Wang. 2004. “Extracellular

HMGB1 as a Proinflammatory Cytokine.” Journal of Interferon & Cytokine Research : The

Official Journal of the International Society for Interferon and Cytokine Research 24 (6):

329–33. doi:10.1089/107999004323142187.

Chiriva-Internati, Maurizio, Fabio Grizzi, Justin Pinkston, K John Morrow, Nicholas D’Cunha,

Eldo E Frezza, Pier Carlo Muzzio, W Martin Kast, and Everardo Cobos. “Gamma-

Radiation Upregulates MHC Class I/II and ICAM-I Molecules in Multiple Myeloma Cell

Lines and Primary Tumors.” In Vitro Cellular & Developmental Biology. Animal 42 (3-4):

89–95. doi:10.1290/0508054.1.

Cho, B C John, Ron Feld, Natasha Leighl, Isabelle Opitz, Masaki Anraku, Ming-Sound Tsao,

David M Hwang, Andrew Hope, and Marc de Perrot. 2014. “A Feasibility Study Evaluating

Surgery for Mesothelioma After Radiation Therapy: The ‘SMART’ Approach for

Resectable Malignant Pleural Mesothelioma.” Journal of Thoracic Oncology : Official

113

Publication of the International Association for the Study of Lung Cancer 9 (3): 397–402.

doi:10.1097/JTO.0000000000000078.

Choe, N., S. Tanaka, W. Xia, D. R. Hemenway, V. L. Roggli, and E. Kagan. 1997. “Pleural

Macrophage Recruitment and Activation in Asbestos-Induced Pleural Injury.”

Environmental Health Perspectives 105 Suppl (September): 1257–60.

doi:10.1289/ehp.97105s51257.

Ciernik, I F, P Romero, J A Berzofsky, and D P Carbone. 1999. “Ionizing Radiation Enhances

Immunogenicity of Cells Expressing a Tumor-Specific T-Cell Epitope.” International

Journal of Radiation Oncology, Biology, Physics 45 (3): 735–41.

http://www.ncbi.nlm.nih.gov/pubmed/10524430.

Clements, Mark, Geoffrey Berry, Jill Shi, Sandra Ware, Deborah Yates, and Anthony Johnson.

2007. “Projected Mesothelioma Incidence in Men in New South Wales.” Occupational and

Environmental Medicine 64 (11): 747–52. doi:10.1136/oem.2006.031823.

Colacchio, T A, M P Yeager, and L W Hildebrandt. 1994. “Perioperative Immunomodulation in

Cancer Surgery.” American Journal of Surgery 167 (1): 174–79.

http://www.ncbi.nlm.nih.gov/pubmed/8311130.

Cree, M, M Lalji, B Jiang, K Carriere, J Beach, and A Kamruzzaman. 2009. “Explaining

Alberta’s Rising Mesothelioma Rates - Chronic Diseases in Canada - Volume 29, No. 4,

2009 - Public Health Agency Canada.” Chronic Diseases in Canada. Government of

Canada, Public Health Agency of Canada. 29 (4): 144–52. http://www.phac-

aspc.gc.ca/publicat/hpcdp-pspmc/29-4/ar_01-eng.php.

Cugell, David W, and David W Kamp. 2004. “Asbestos and the Pleura: A Review.” Chest 125

(3): 1103–17. http://www.ncbi.nlm.nih.gov/pubmed/15006974.

Currie, Andrew J, Amy Prosser, Alison McDonnell, Amanda L Cleaver, Bruce W S Robinson,

Gordon J Freeman, and Robbert G van der Most. 2009. “Dual Control of Antitumor CD8 T

Cells through the Programmed Death-1/programmed Death-Ligand 1 Pathway and

Immunosuppressive CD4 T Cells: Regulation and Counterregulation.” Journal of

Immunology (Baltimore, Md. : 1950) 183 (12): 7898–7908. doi:10.4049/jimmunol.0901060.

114

Dąbrowska, Aleksandra M., and Robert Słotwiński. 2014. “The Immune Response to Surgery

and Infection.” Central European Journal of Immunology 4 (4): 532–37.

doi:10.5114/ceji.2014.47741.

Davila, Eduardo, Richard Kennedy, and Esteban Celis. 2003. “Generation of Antitumor

Immunity by Cytotoxic T Lymphocyte Epitope Peptide Vaccination, CpG-

Oligodeoxynucleotide Adjuvant, and CTLA-4 Blockade.” Cancer Research 63 (12): 3281–

88. http://www.ncbi.nlm.nih.gov/pubmed/12810660.

De Wever, Olivier, and Marc Mareel. 2003. “Role of Tissue Stroma in Cancer Cell Invasion.”

The Journal of Pathology 200 (4): 429–47. doi:10.1002/path.1398.

Demaria, Sandra, Nina Bhardwaj, William H McBride, and Silvia C Formenti. 2005.

“Combining Radiotherapy and Immunotherapy: A Revived Partnership.” International

Journal of Radiation Oncology, Biology, Physics 63 (3): 655–66.

doi:10.1016/j.ijrobp.2005.06.032.

Demaria, Sandra, and Silvia C. Formenti. 2012. “Radiation as an Immunological Adjuvant:

Current Evidence on Dose and Fractionation.” Frontiers in Oncology 2 (October): 1–7.

doi:10.3389/fonc.2012.00153.

Demaria, Sandra, Noriko Kawashima, Anne Marie Yang, Mary Louise Devitt, James S. Babb,

James P. Allison, and Silvia C. Formenti. 2005. “Immune-Mediated Inhibition of

Metastases after Treatment with Local Radiation and CTLA-4 Blockade in a Mouse Model

of Breast Cancer.” Clinical Cancer Research 11 (2 I): 728–34. doi:11/2/728 [pii].

Demaria, Sandra, Bruce Ng, Mary Louise Devitt, James S. Babb, Noriko Kawashima, Leonard

Liebes, and Silvia C. Formenti. 2004. “Ionizing Radiation Inhibition of Distant Untreated

Tumors (abscopal Effect) Is Immune Mediated.” International Journal of Radiation

Oncology Biology Physics 58 (3): 862–70. doi:10.1016/j.ijrobp.2003.09.012.

Demicheli, R, M W Retsky, W J M Hrushesky, M Baum, and I D Gukas. 2008. “The Effects of

Surgery on Tumor Growth: A Century of Investigations.” Annals of Oncology : Official

Journal of the European Society for Medical Oncology / ESMO 19 (11): 1821–28.

doi:10.1093/annonc/mdn386.

115

Demirag, Funda, Ebru Unsal, Aydin Yilmaz, and Atalay Caglar. 2005. “Prognostic Significance

of Vascular Endothelial Growth Factor, Tumor Necrosis, and Mitotic Activity Index in

Malignant Pleural Mesothelioma.” Chest 128 (5): 3382–87. doi:10.1378/chest.128.5.3382.

Desai, Sejal, Amit Kumar, S Laskar, and B N Pandey. 2013. “Cytokine Profile of Conditioned

Medium from Human Tumor Cell Lines after Acute and Fractionated Doses of Gamma

Radiation and Its Effect on Survival of Bystander Tumor Cells.” Cytokine 61 (1): 54–62.

doi:10.1016/j.cyto.2012.08.022.

Desborough, J P. 2000. “The Stress Response to Trauma and Surgery.” British Journal of

Anaesthesia 85 (1): 109–17. http://www.ncbi.nlm.nih.gov/pubmed/10927999.

Dewan, M. Zahidunnabi, Ashley E. Galloway, Noriko Kawashima, J. Keith Dewyngaert, James

S. Babb, Silvia C. Formenti, and Sandra Demaria. 2009. “Fractionated but Not Single-Dose

Radiotherapy Induces an Immune-Mediated Abscopal Effect When Combined with Anti-

CTLA-4 Antibody.” Clinical Cancer Research 15 (17): 5379–88. doi:10.1158/1078-

0432.CCR-09-0265.

Dhabhar, F S, A H Miller, B S McEwen, and R L Spencer. 1996. “Stress-Induced Changes in

Blood Leukocyte Distribution. Role of Adrenal Steroid Hormones.” Journal of Immunology

(Baltimore, Md. : 1950) 157 (4): 1638–44. http://www.ncbi.nlm.nih.gov/pubmed/8759750.

Di Stasi, Antonio, Siok-Keen Tey, Gianpietro Dotti, Yuriko Fujita, Alana Kennedy-Nasser,

Caridad Martinez, Karin Straathof, et al. 2011. “Inducible Apoptosis as a Safety Switch for

Adoptive Cell Therapy.” The New England Journal of Medicine 365 (18): 1673–83.

doi:10.1056/NEJMoa1106152.

Dighe, A S, E Richards, L J Old, and R D Schreiber. 1994. “Enhanced in Vivo Growth and

Resistance to Rejection of Tumor Cells Expressing Dominant Negative IFN Gamma

Receptors.” Immunity 1 (6): 447–56. http://www.ncbi.nlm.nih.gov/pubmed/7895156.

Dowell, Jonathan E, Frank R Dunphy, Robert N Taub, David E Gerber, Likheng Ngov,

Jingsheng Yan, Yang Xie, and Hedy Lee Kindler. 2012. “A Multicenter Phase II Study of

Cisplatin, Pemetrexed, and Bevacizumab in Patients with Advanced Malignant

116

Mesothelioma.” Lung Cancer (Amsterdam, Netherlands) 77 (3): 567–71.

doi:10.1016/j.lungcan.2012.05.111.

Draghiciu, Oana, Mateusz Walczak, Baukje Nynke Hoogeboom, Kees L M C Franken, Kees J M

Melief, Hans W Nijman, and Toos Daemen. 2014. “Therapeutic Immunization and Local

Low-Dose Tumor Irradiation, a Reinforcing Combination.” International Journal of

Cancer. Journal International Du Cancer 134 (4): 859–72. doi:10.1002/ijc.28418.

Driece, Hermen a L, Sabine Siesling, Paul H J J Swuste, and Alex Burdorf. 2010. “Assessment

of Cancer Risks due to Environmental Exposure to Asbestos.” Journal of Exposure Science

& Environmental Epidemiology 20 (5). Nature Publishing Group: 478–85.

doi:10.1038/jes.2009.56.

Driessens, Gregory, Justin Kline, and Thomas F. Gajewski. 2009. “Costimulatory and

Coinhibitory Receptors in Anti-Tumor Immunity.” Immunological Reviews 229 (1): 126–

44. doi:10.1111/j.1600-065X.2009.00771.x.

Dudley, Mark E, John R Wunderlich, Paul F Robbins, James C Yang, Patrick Hwu, Douglas J

Schwartzentruber, Suzanne L Topalian, et al. 2002. “Cancer Regression and Autoimmunity

in Patients after Clonal Repopulation with Antitumor Lymphocytes.” Science (New York,

N.Y.) 298 (5594): 850–54. doi:10.1126/science.1076514.

Dunn, Gavin P, Allen T Bruce, Hiroaki Ikeda, Lloyd J Old, and Robert D Schreiber. 2002.

“Cancer Immunoediting: From Immunosurveillance to Tumor Escape.” Nature Immunology

3 (11): 991–98. doi:10.1038/ni1102-991.

Ehrlich, Paul. 1909. “Über Den Jetzigen Stand Der Karzinomforschung.” Ned. Tijdschr.

Geneeskd. 5: 273–90.

Elenkov, I J, R L Wilder, G P Chrousos, and E S Vizi. 2000. “The Sympathetic Nerve--an

Integrative Interface between Two Supersystems: The Brain and the Immune System.”

Pharmacological Reviews 52 (4): 595–638.

http://www.ncbi.nlm.nih.gov/pubmed/11121511.

117

Elenkov, Ilia J. 2004. “Glucocorticoids and the Th1/Th2 Balance.” Annals of the New York

Academy of Sciences 1024 (1): 138–46. doi:10.1196/annals.1321.010.

Eriksson, David, and Torgny Stigbrand. 2010. “Radiation-Induced Cell Death Mechanisms.”

Tumour Biology : The Journal of the International Society for Oncodevelopmental Biology

and Medicine 31 (4): 363–72. doi:10.1007/s13277-010-0042-8.

Ettinger, D S, W Akerley, H Borghaei, a Chang, R T Cheney, L R Chirieac, T a D’Amico, et al.

2012. “Malignant Pleural Mesothelioma. Clinical Practice Guidelines in Oncology.” J Natl

Compr Canc Netw 10 (1): 26–41. doi:10/1/26 [pii].

Fedorocko, P, A Egyed, and A Vacek. 2002. “Irradiation Induces Increased Production of

Haemopoietic and Proinflammatory Cytokines in the Mouse Lung.” International Journal

of Radiation Biology 78 (4): 305–13. doi:10.1080/09553000110104614.

Filatenkov, Alexander, Jeanette Baker, Antonia M S Mueller, Justin Kenkel, G-One Ahn,

Suparna Dutt, Nigel Zhang, et al. 2015. “Ablative Tumor Radiation Can Change the Tumor

Immune Cell Microenvironment to Induce Durable Complete Remissions.” Clinical Cancer

Research : An Official Journal of the American Association for Cancer Research, April.

doi:10.1158/1078-0432.CCR-14-2824.

Fisher, S G, L Weber, and M Carbone. “Cancer Risk Associated with Simian Virus 40

Contaminated Polio Vaccine.” Anticancer Research 19 (3B): 2173–80.

http://www.ncbi.nlm.nih.gov/pubmed/10472327.

Fourcade, Julien, Zhaojun Sun, Mourad Benallaoua, Philippe Guillaume, Immanuel F Luescher,

Cindy Sander, John M Kirkwood, Vijay Kuchroo, and Hassane M Zarour. 2010.

“Upregulation of Tim-3 and PD-1 Expression Is Associated with Tumor Antigen-Specific

CD8+ T Cell Dysfunction in Melanoma Patients.” The Journal of Experimental Medicine

207 (10): 2175–86. doi:10.1084/jem.20100637.

Frey, Benjamin, Yvonne Rubner, Lorenz Kulzer, Nina Werthmöller, Eva-Maria Weiss, Rainer

Fietkau, and Udo S Gaipl. 2014. “Antitumor Immune Responses Induced by Ionizing

Irradiation and Further Immune Stimulation.” Cancer Immunology, Immunotherapy : CII 63

(1): 29–36. doi:10.1007/s00262-013-1474-y.

118

Fuhrer, Gregory, and Angeline a. Lazarus. 2011. “Mesothelioma.” Disease-a-Month 57 (1): 40–

54. doi:10.1016/j.disamonth.2010.12.004.

Gajewski, Thomas F, Mercedes Fuertes, Robbert Spaapen, Yan Zheng, and Justin Kline. 2011.

“Molecular Profiling to Identify Relevant Immune Resistance Mechanisms in the Tumor

Microenvironment.” Current Opinion in Immunology 23 (2): 286–92.

doi:10.1016/j.coi.2010.11.013.

Gao, Y, J M Herndon, H Zhang, T S Griffith, and T A Ferguson. 1998. “Antiinflammatory

Effects of CD95 Ligand (FasL)-Induced Apoptosis.” The Journal of Experimental Medicine

188 (5): 887–96.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2213381&tool=pmcentrez&ren

dertype=abstract.

Garg, Abhishek D, Dmitri V Krysko, Tom Verfaillie, Agnieszka Kaczmarek, Gabriela B

Ferreira, Thierry Marysael, Noemi Rubio, et al. 2012. “A Novel Pathway Combining

Calreticulin Exposure and ATP Secretion in Immunogenic Cancer Cell Death.” The EMBO

Journal 31 (5): 1062–79. doi:10.1038/emboj.2011.497.

Garland, Linda L, Cathryn Rankin, David R Gandara, Saul E Rivkin, Katherine M Scott,

Raymond B Nagle, Andres J P Klein-Szanto, Joseph R Testa, Deborah A Altomare, and

Ernest C Borden. 2007. “Phase II Study of Erlotinib in Patients with Malignant Pleural

Mesothelioma: A Southwest Oncology Group Study.” Journal of Clinical Oncology :

Official Journal of the American Society of Clinical Oncology 25 (17): 2406–13.

doi:10.1200/JCO.2006.09.7634.

Garland, Linda L. 2011. “Chemotherapy for Malignant Pleural Mesothelioma.” Current

Treatment Options in Oncology 12 (2): 181–88. doi:10.1007/s11864-011-0152-6.

Garnett, Charlie T, Claudia Palena, Mala Chakarborty, Kwong-yok Tsang, Jeffrey Schlom, and

James W Hodge. 2004. “Sublethal Irradiation of Human Tumor Cells Modulates Phenotype

Resulting in Enhanced Killing by Cytotoxic T Lymphocytes.” Cancer Research 64: 7985–

94.

119

Gazdar, Adi F, Janet S Butel, and Michele Carbone. 2002. “SV40 and Human Tumours: Myth,

Association or Causality?” Nature Reviews. Cancer 2 (12): 957–64. doi:10.1038/nrc947.

Ghiringhelli, François, Lionel Apetoh, Antoine Tesniere, Laetitia Aymeric, Yuting Ma, Carla

Ortiz, Karim Vermaelen, et al. 2009. “Activation of the NLRP3 Inflammasome in Dendritic

Cells Induces IL-1beta-Dependent Adaptive Immunity against Tumors.” Nature Medicine

15 (10): 1170–78. doi:10.1038/nm.2028.

Golden, Encouse B, and Lionel Apetoh. 2015. “Radiotherapy and Immunogenic Cell Death.”

Seminars in Radiation Oncology 25 (1): 11–17. doi:10.1016/j.semradonc.2014.07.005.

Golden, Encouse B, Sandra Demaria, Peter B Schiff, Abraham Chachoua, and Silvia C Formenti.

2013. “An Abscopal Response to Radiation and Ipilimumab in a Patient with Metastatic

Non-Small Cell Lung Cancer.” Cancer Immunology Research 1 (6): 365–72.

doi:10.1158/2326-6066.CIR-13-0115.

Golden, Encouse B., Ilenia Pellicciotta, Sandra Demaria, Mary H. Barcellos-Hoff, and Silvia C.

Formenti. 2012. “The Convergence of Radiation and Immunogenic Cell Death Signaling

Pathways.” Frontiers in Oncology 2 (August): 1–13. doi:10.3389/fonc.2012.00088.

Gottlieb, Tanya M, Juan Fernando Martinez Leal, Rony Seger, Yoichi Taya, and Moshe Oren.

2002. “Cross-Talk between Akt, p53 and Mdm2: Possible Implications for the Regulation

of Apoptosis.” Oncogene 21 (8): 1299–1303. doi:10.1038/sj.onc.1205181.

Govindan, Ramaswamy, Robert A Kratzke, James E Herndon, Gloria A Niehans, Robin

Vollmer, Dorothy Watson, Mark R Green, and Hedy L Kindler. 2005. “Gefitinib in Patients

with Malignant Mesothelioma: A Phase II Study by the Cancer and Leukemia Group B.”

Clinical Cancer Research : An Official Journal of the American Association for Cancer

Research 11 (6): 2300–2304. doi:10.1158/1078-0432.CCR-04-1940.

Gross, G, G Gorochov, T Waks, and Z Eshhar. 1989. “Generation of Effector T Cells Expressing

Chimeric T Cell Receptor with Antibody Type-Specificity.” Transplantation Proceedings

21 (1 Pt 1): 127–30. http://www.ncbi.nlm.nih.gov/pubmed/2784887.

120

Grosso, Joseph F., and Maria N. Jure-Kunkel. 2013. “CTLA-4 Blockade in Tumor Models: An

Overview of Preclinical and Translational Research.” Cancer Immunity 13 (January): 5.

Gupta, Anurag, Hans Christian Probst, Van Vuong, Alexandro Landshammer, Sabine Muth,

Hideo Yagita, Reto Schwendener, Martin Pruschy, Alexander Knuth, and Maries van den

Broek. 2012. “Radiotherapy Promotes Tumor-Specific Effector CD8+ T Cells via Dendritic

Cell Activation.” Journal of Immunology (Baltimore, Md. : 1950) 189 (2): 558–66.

doi:10.4049/jimmunol.1200563.

Hall, Eric J., and Amato J. Giaccia. 2006. “Physics and Chemistry of Radiation Absorption.” In

Radiobiology for the Radiologist, 6th ed., 5–15. Lippincott Williams & Wilkins.

Hallahan, D, J Kuchibhotla, and C Wyble. 1996. “Cell Adhesion Molecules Mediate Radiation-

Induced Leukocyte Adhesion to the Vascular Endothelium.” Cancer Research 56 (22):

5150–55. http://www.ncbi.nlm.nih.gov/pubmed/8912850.

Hamdi, Mohamed, Jaap Kool, Paulien Cornelissen-Steijger, Francoise Carlotti, Herman E

Popeijus, Corina van der Burgt, Josephine M Janssen, et al. 2005. “DNA Damage in

Transcribed Genes Induces Apoptosis via the JNK Pathway and the JNK-Phosphatase

MKP-1.” Oncogene 24 (48): 7135–44. doi:10.1038/sj.onc.1208875.

Hamid, Omid, Caroline Robert, Adil Daud, F Stephen Hodi, Wen-Jen Hwu, Richard Kefford,

Jedd D Wolchok, et al. 2013. “Safety and Tumor Responses with Lambrolizumab (anti-PD-

1) in Melanoma.” The New England Journal of Medicine 369 (2): 134–44.

doi:10.1056/NEJMoa1305133.

Hareyama, M, K Imai, K Kubo, H Takahashi, H Koshiba, Y Hinoda, M Shidou, A Oouchi, A

Yachi, and K Morita. 1991. “Effect of Radiation on the Expression of Carcinoembryonic

Antigen of Human Gastric Adenocarcinoma Cells.” Cancer 67 (9): 2269–74.

http://www.ncbi.nlm.nih.gov/pubmed/1901513.

Harrison, L H, P O Schwarzenberger, P S Byrne, A J Marrogi, J K Kolls, and K E McCarthy.

2000. “Gene-Modified PA1-STK Cells Home to Tumor Sites in Patients with Malignant

Pleural Mesothelioma.” The Annals of Thoracic Surgery 70 (2): 407–11.

http://www.ncbi.nlm.nih.gov/pubmed/10969653.

121

Hassan, Raffit, Steven J Cohen, Martin Phillips, Ira Pastan, Elad Sharon, Ronan J Kelly, Charles

Schweizer, Susan Weil, and Daniel Laheru. 2010. “Phase I Clinical Trial of the Chimeric

Anti-Mesothelin Monoclonal Antibody MORAb-009 in Patients with Mesothelin-

Expressing Cancers.” Clinical Cancer Research : An Official Journal of the American

Association for Cancer Research 16 (24): 6132–38. doi:10.1158/1078-0432.CCR-10-2275.

Hassan, Raffit, Wolfgang Ebel, Eric L Routhier, Rina Patel, J Bradford Kline, Jingli Zhang,

Qimin Chao, et al. 2007. “Preclinical Evaluation of MORAb-009, a Chimeric Antibody

Targeting Tumor-Associated Mesothelin.” Cancer Immunity 7 (January): 20.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2935758&tool=pmcentrez&ren

dertype=abstract.

Hassan, Raffit, Hedy L Kindler, Thierry Jahan, Lyudmila Bazhenova, Martin Reck, Anish

Thomas, Ira Pastan, et al. 2014. “Phase II Clinical Trial of Amatuximab, a Chimeric

Antimesothelin Antibody with Pemetrexed and Cisplatin in Advanced Unresectable Pleural

Mesothelioma.” Clinical Cancer Research : An Official Journal of the American

Association for Cancer Research 20 (23): 5927–36. doi:10.1158/1078-0432.CCR-14-0804.

Hassan, Raffit, Andrew C Miller, Elad Sharon, Anish Thomas, James C Reynolds, Alexander

Ling, Robert J Kreitman, et al. 2013. “Major Cancer Regressions in Mesothelioma after

Treatment with an Anti-Mesothelin Immunotoxin and Immune Suppression.” Science

Translational Medicine 5 (208): 208ra147. doi:10.1126/scitranslmed.3006941.

Haupt, Susan, Michael Berger, Zehavit Goldberg, and Ygal Haupt. 2003. “Apoptosis - the p53

Network.” Journal of Cell Science 116 (Pt 20): 4077–85. doi:10.1242/jcs.00739.

Hegmans, Joost P, Joris D Veltman, Margaretha E Lambers, I Jolanda M de Vries, Carl G

Figdor, Rudi W Hendriks, Henk C Hoogsteden, Bart N Lambrecht, and Joachim G Aerts.

2010. “Consolidative Dendritic Cell-Based Immunotherapy Elicits Cytotoxicity against

Malignant Mesothelioma.” American Journal of Respiratory and Critical Care Medicine

181 (12): 1383–90. doi:10.1164/rccm.200909-1465OC.

122

Heiblig, Maël, Mohamed Elhamri, Mauricette Michallet, and Xavier Thomas. 2015. “Adoptive

Immunotherapy for Acute Leukemia: New Insights in Chimeric Antigen Receptors.” World

Journal of Stem Cells 7 (7): 1022–38. doi:10.4252/wjsc.v7.i7.1022.

Heinonen, O P, S Shapiro, R R Monson, S C Hartz, L Rosenberg, and D Slone. 1973.

“Immunization during Pregnancy against Poliomyelitis and Influenza in Relation to

Childhood Malignancy.” International Journal of Epidemiology 2 (3): 229–35.

http://www.ncbi.nlm.nih.gov/pubmed/4359832.

Henkart, P A. 1994. “Lymphocyte-Mediated Cytotoxicity: Two Pathways and Multiple Effector

Molecules.” Immunity 1 (5): 343–46. http://www.ncbi.nlm.nih.gov/pubmed/7882166.

Hernandez-Chacon, Jessica Ann, Yufeng Li, Richard C Wu, Chantale Bernatchez, Yijun Wang,

Jeffrey S Weber, Patrick Hwu, and Laszlo G Radvanyi. 2011. “Costimulation through the

CD137/4-1BB Pathway Protects Human Melanoma Tumor-Infiltrating Lymphocytes from

Activation-Induced Cell Death and Enhances Antitumor Effector Function.” Journal of

Immunotherapy (Hagerstown, Md. : 1997) 34 (3): 236–50.

doi:10.1097/CJI.0b013e318209e7ec.

Heylmann, Daniel, Franz Rödel, Thomas Kindler, and Bernd Kaina. 2014. “Radiation Sensitivity

of Human and Murine Peripheral Blood Lymphocytes, Stem and Progenitor Cells.”

Biochimica et Biophysica Acta 1846 (1): 121–29. doi:10.1016/j.bbcan.2014.04.009.

Hill, R J, R E Edwards, and P Carthew. 1990. “Early Changes in the Pleural Mesothelium

Following Intrapleural Inoculation of the Mineral Fibre Erionite and the Subsequent

Development of Mesotheliomas.” Journal of Experimental Pathology (Oxford, England) 71

(1): 105–18.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1998670&tool=pmcentrez&ren

dertype=abstract.

Hirte, Hal, and David A. Clark. 1991. “Generation of Lymphokine-Activated Killer Cells in

Human Ovarian Carcinoma Ascitic Fluid: Identification of Transforming Growth Factor-Β

as a Suppressive Factor.” Cancer Immunology Immunotherapy 32 (5): 296–302.

doi:10.1007/BF01789047.

123

Hodgson, J T, and A Darnton. 2000. “The Quantitative Risks of Mesothelioma and Lung Cancer

in Relation to Asbestos Exposure.” The Annals of Occupational Hygiene 44 (8): 565–601.

http://www.ncbi.nlm.nih.gov/pubmed/11108782.

Hodgson, J T, D M McElvenny, A J Darnton, M J Price, and J Peto. 2005. “The Expected

Burden of Mesothelioma Mortality in Great Britain from 2002 to 2050.” British Journal of

Cancer 92 (3): 587–93. doi:10.1038/sj.bjc.6602307.

Hodi, F Stephen, Steven J O’Day, David F McDermott, Robert W Weber, Jeffrey A Sosman,

John B Haanen, Rene Gonzalez, et al. 2010. “Improved Survival with Ipilimumab in

Patients with Metastatic Melanoma.” The New England Journal of Medicine 363 (8): 711–

23. doi:10.1056/NEJMoa1003466.

Hofer, S O, G Molema, R A Hermens, H J Wanebo, J S Reichner, and H J Hoekstra. 1999. “The

Effect of Surgical Wounding on Tumour Development.” European Journal of Surgical

Oncology : The Journal of the European Society of Surgical Oncology and the British

Association of Surgical Oncology 25 (3): 231–43. doi:10.1053/ejso.1998.0634.

Hofer, S O, D Shrayer, J S Reichner, H J Hoekstra, and H J Wanebo. 1998. “Wound-Induced

Tumor Progression: A Probable Role in Recurrence after Tumor Resection.” Archives of

Surgery (Chicago, Ill. : 1960) 133 (4): 383–89.

http://www.ncbi.nlm.nih.gov/pubmed/9565118.

Hormbrey, Emma, Cheng Han, Anthony Roberts, Duncan Angus McGrouther, and Adrian L

Harris. 2003. “The Relationship of Human Wound Vascular Endothelial Growth Factor

(VEGF) after Breast Cancer Surgery to Circulating VEGF and Angiogenesis.” Clinical

Cancer Research : An Official Journal of the American Association for Cancer Research 9

(12): 4332–39. http://www.ncbi.nlm.nih.gov/pubmed/14555503.

Hsu, Elisabeth. 2006. “The History of.” In Tropical Medicine, edited by Harvey I. Pass, Nicholas

J. Vogelzang, and Michele Carbone, 1st ed., 3–20. New York, NY: Springer.

doi:10.1016/j.trstmh.2005.09.020.

Illidge, T M. 1998. “Radiation-Induced Apoptosis.” Clinical Oncology (Royal College of

Radiologists (Great Britain)) 10 (1): 3–13. http://www.ncbi.nlm.nih.gov/pubmed/9543608.

124

Innis, M D. 1968. “Oncogenesis and Poliomyelitis Vaccine.” Nature 219 (5157): 972–73.

http://www.ncbi.nlm.nih.gov/pubmed/4299754.

Ismail-Khan, Roohi, Lary a Robinson, Charles C Williams, Christopher R Garrett, Gerold

Bepler, and George R Simon. 2006. “Malignant Pleural Mesothelioma: A Comprehensive

Review.” Cancer Control : Journal of the Moffitt Cancer Center 13 (4): 255–63.

Jackaman, Connie, Christine S Bundell, Beverley F Kinnear, Alison M Smith, Pierre Filion,

Deborah van Hagen, Bruce W S Robinson, and Delia J Nelson. 2003. “IL-2 Intratumoral

Immunotherapy Enhances CD8+ T Cells That Mediate Destruction of Tumor Cells and

Tumor-Associated Vasculature: A Novel Mechanism for IL-2.” Journal of Immunology

(Baltimore, Md. : 1950) 171 (10): 5051–63. doi:10.4049/jimmunol.171.10.5051.

Jahan, Thierry, Lin Gu, Robert Kratzke, Arkadiusz Dudek, Gregory A Otterson, Xiaofei Wang,

Mark Green, Everett E Vokes, and Hedy Lee Kindler. 2012. “Vatalanib in Malignant

Mesothelioma: A Phase II Trial by the Cancer and Leukemia Group B (CALGB 30107).”

Lung Cancer (Amsterdam, Netherlands) 76 (3): 393–96.

doi:10.1016/j.lungcan.2011.11.014.

Jameson, P, J P Desborough, A E Bryant, and G M Hall. 1997. “The Effect of Cortisol

Suppression on Interleukin-6 and White Blood Cell Responses to Surgery.” Acta

Anaesthesiologica Scandinavica 41 (2): 304–8.

http://www.ncbi.nlm.nih.gov/pubmed/9062617.

Jänne, Pasi A, Michele L Taffaro, Ravi Salgia, and Bruce E Johnson. 2002. “Inhibition of

Epidermal Growth Factor Receptor Signaling in Malignant Pleural Mesothelioma.” Cancer

Research 62 (18): 5242–47. http://www.ncbi.nlm.nih.gov/pubmed/12234991.

Jasani, B, A Cristaudo, S A Emri, A F Gazdar, A Gibbs, B Krynska, C Miller, et al. 2001.

“Association of SV40 with Human Tumours.” Seminars in Cancer Biology 11 (1): 49–61.

doi:10.1006/scbi.2000.0346.

Jonathan, E C, E J Bernhard, and W G McKenna. 1999. “How Does Radiation Kill Cells?”

Current Opinion in Chemical Biology 3 (1): 77–83.

http://www.ncbi.nlm.nih.gov/pubmed/10021401.

125

Kaina, Bernd, Wynand Roos, and Markus Christmann. 2010. “DNA Damage Response and the

Balance Between Cell Survival and Cell Death.” In Checkpoint Controls and Targets in

Cancer Therapy, edited by Zahid H. Siddik, First, 95–108. Springer Science & Business

Media.

Kal, Henk B, Henk Struikmans, and Angelique D Barten-van Rijbroek. 2008. “Surgical Stress

and Accelerated Tumor Growth.” Anticancer Research 28 (2A): 1129–32.

http://www.ncbi.nlm.nih.gov/pubmed/18507064.

Kalbasi, Anusha, Carl H. June, Naomi Haas, and Neha Vapiwala. 2013. “Radiation and

Immunotherapy: A Synergistic Combination.” Journal of Clinical Investigation.

doi:10.1172/JCI69219.

Kamp, D W, V A Israbian, S E Preusen, C X Zhang, and S A Weitzman. 1995. “Asbestos

Causes DNA Strand Breaks in Cultured Pulmonary Epithelial Cells: Role of Iron-Catalyzed

Free Radicals.” The American Journal of Physiology 268 (3 Pt 1): L471–80.

http://www.ncbi.nlm.nih.gov/pubmed/7900829.

Kaplan, H S. 1971. “Role of Immunologic Disturbance in Human Oncogenesis: Some Facts and

Fancies.” British Journal of Cancer 25 (4): 620–34.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2008860&tool=pmcentrez&ren

dertype=abstract.

Karin, M, and Y Ben-Neriah. 2000. “Phosphorylation Meets Ubiquitination: The Control of NF-

[kappa]B Activity.” Annual Review of Immunology 18 (January): 621–63.

doi:10.1146/annurev.immunol.18.1.621.

Kehlet, H. 1997. “Multimodal Approach to Control Postoperative Pathophysiology and

Rehabilitation.” British Journal of Anaesthesia 78 (5): 606–17.

http://www.ncbi.nlm.nih.gov/pubmed/9175983.

Keir, Mary E, Manish J Butte, Gordon J Freeman, and Arlene H Sharpe. 2008. “PD-1 and Its

Ligands in Tolerance and Immunity.” Annual Review of Immunology 26 (January): 677–

704. doi:10.1146/annurev.immunol.26.021607.090331.

126

Keisari, Yona, Ilan Hochman, Hila Confino, Rafi Korenstein, and Itzhak Kelson. 2014.

“Activation of Local and Systemic Anti-Tumor Immune Responses by Ablation of Solid

Tumors with Intratumoral Electrochemical or Alpha Radiation Treatments.” Cancer

Immunology, Immunotherapy 63 (1): 1–9. doi:10.1007/s00262-013-1462-2.

Kepp, Oliver, Lorenzo Galluzzi, Isabelle Martins, Frederic Schlemmer, Sandy Adjemian,

Mickael Michaud, Abdul Qader Sukkurwala, Laurie Menger, Laurence Zitvogel, and Guido

Kroemer. 2011. “Molecular Determinants of Immunogenic Cell Death Elicited by

Anticancer Chemotherapy.” Cancer Metastasis Reviews 30 (1): 61–69. doi:10.1007/s10555-

011-9273-4.

Kershaw, Michael H, Jennifer A Westwood, Linda L Parker, Gang Wang, Zelig Eshhar, Sharon

A Mavroukakis, Donald E White, et al. 2006. “A Phase I Study on Adoptive

Immunotherapy Using Gene-Modified T Cells for Ovarian Cancer.” Clinical Cancer

Research : An Official Journal of the American Association for Cancer Research 12 (20 Pt

1): 6106–15. doi:10.1158/1078-0432.CCR-06-1183.

Kim, Ryungsa, Manabu Emi, and Kazuaki Tanabe. 2007. “Cancer Immunoediting from Immune

Surveillance to Immune Escape.” Immunology 121 (1): 1–14. doi:10.1111/j.1365-

2567.2007.02587.x.

Kim, Sunhwa, Hiroyuki Takahashi, Wan-Wan Lin, Pascal Descargues, Sergei Grivennikov,

Youngjun Kim, Jun-Li Luo, and Michael Karin. 2009. “Carcinoma-Produced Factors

Activate Myeloid Cells through TLR2 to Stimulate Metastasis.” Nature 457 (7225): 102–6.

doi:10.1038/nature07623.

Kimura, Fumio, Hiroaki Shimizu, Hiroyuki Yoshidome, Masayuki Ohtsuka, and Masaru

Miyazaki. 2010. “Immunosuppression Following Surgical and Traumatic Injury.” Surgery

Today 40 (9): 793–808. doi:10.1007/s00595-010-4323-z.

Klug, Felix, Hridayesh Prakash, Peter E Huber, Tobias Seibel, Noemi Bender, Niels Halama,

Christina Pfirschke, et al. 2013. “Low-Dose Irradiation Programs Macrophage

Differentiation to an iNOS+/M1 Phenotype That Orchestrates Effective T Cell

Immunotherapy.” Cancer Cell 24 (5): 589–602. doi:10.1016/j.ccr.2013.09.014.

127

König, J, E Tolnay, T Wiethege, and K Müller. 2000. “Co-Expression of Vascular Endothelial

Growth Factor and Its Receptor Flt-1 in Malignant Pleural Mesothelioma.” Respiration;

International Review of Thoracic Diseases 67 (1): 36–40. doi:29460.

Kono, Hajime, and Kenneth L Rock. 2008. “How Dying Cells Alert the Immune System to

Danger.” Nature Reviews. Immunology 8 (4): 279–89. doi:10.1038/nri2215.

Kotova, Svetlana, Raymond M Wong, and Robert B Cameron. 2015. “New and Emerging

Therapeutic Options for Malignant Pleural Mesothelioma : Review of Early Clinical Trials.”

Cancer Management and Research 7: 51–63.

Kreitman, Robert J, Raffit Hassan, David J Fitzgerald, and Ira Pastan. 2009. “Phase I Trial of

Continuous Infusion Anti-Mesothelin Recombinant Immunotoxin SS1P.” Clinical Cancer

Research : An Official Journal of the American Association for Cancer Research 15 (16):

5274–79. doi:10.1158/1078-0432.CCR-09-0062.

Kroemer, Guido, Lorenzo Galluzzi, Oliver Kepp, and Laurence Zitvogel. 2013. “Immunogenic

Cell Death in Cancer Therapy.” Annual Review of Immunology 31 (January): 51–72.

doi:10.1146/annurev-immunol-032712-100008.

Krug, Lee M, Tao Dao, Andrew B Brown, Peter Maslak, William Travis, Sara Bekele, Tatyana

Korontsvit, et al. 2010. “WT1 Peptide Vaccinations Induce CD4 and CD8 T Cell Immune

Responses in Patients with Mesothelioma and Non-Small Cell Lung Cancer.” Cancer

Immunology, Immunotherapy : CII 59 (10): 1467–79. doi:10.1007/s00262-010-0871-8.

Krysko, O, T Løve Aaes, C Bachert, P Vandenabeele, and D V Krysko. 2013. “Many Faces of

DAMPs in Cancer Therapy.” Cell Death & Disease 4 (January): e631.

doi:10.1038/cddis.2013.156.

Krysko, Dmitri V, Abhishek D Garg, Agnieszka Kaczmarek, Olga Krysko, Patrizia Agostinis,

and Peter Vandenabeele. 2012. “Immunogenic Cell Death and DAMPs in Cancer Therapy.”

Nature Reviews. Cancer 12 (12): 860–75. doi:10.1038/nrc3380.

Lamers, Cor Hj, Stefan Sleijfer, Sabine van Steenbergen, Pascal van Elzakker, Brigitte van

Krimpen, Corrien Groot, Arnold Vulto, et al. 2013. “Treatment of Metastatic Renal Cell

128

Carcinoma with CAIX CAR-Engineered T Cells: Clinical Evaluation and Management of

on-Target Toxicity.” Molecular Therapy : The Journal of the American Society of Gene

Therapy 21 (4): 904–12. doi:10.1038/mt.2013.17.

Larkin, James, Vanna Chiarion-Sileni, Rene Gonzalez, Jean Jacques Grob, C Lance Cowey,

Christopher D Lao, Dirk Schadendorf, et al. 2015. “Combined Nivolumab and Ipilimumab

or Monotherapy in Untreated Melanoma.” The New England Journal of Medicine 373 (1):

23–34. doi:10.1056/NEJMoa1504030.

Lauber, Kirsten, Anne Ernst, Michael Orth, Martin Herrmann, and Claus Belka. 2012. “Dying

Cell Clearance and Its Impact on the Outcome of Tumor Radiotherapy.” Frontiers in

Oncology 2 (January): 116. doi:10.3389/fonc.2012.00116.

Lavin, M F. 2007. “ATM and the Mre11 Complex Combine to Recognize and Signal DNA

Double-Strand Breaks.” Oncogene 26 (56): 7749–58. doi:10.1038/sj.onc.1210880.

Le, Dung T, Dirk G Brockstedt, Ran Nir-Paz, Johannes Hampl, Shruti Mathur, John Nemunaitis,

Daniel H Sterman, et al. 2012. “A Live-Attenuated Listeria Vaccine (ANZ-100) and a Live-

Attenuated Listeria Vaccine Expressing Mesothelin (CRS-207) for Advanced Cancers:

Phase I Studies of Safety and Immune Induction.” Clinical Cancer Research : An Official

Journal of the American Association for Cancer Research 18 (3): 858–68.

doi:10.1158/1078-0432.CCR-11-2121.

Lee, Hyeon-Woo, Su-Jung Park, Beom K Choi, Hyun Hwa Kim, Kyung-Ok Nam, and Byoung S

Kwon. 2002. “4-1BB Promotes the Survival of CD8+ T Lymphocytes by Increasing

Expression of Bcl-xL and Bfl-1.” Journal of Immunology (Baltimore, Md. : 1950) 169 (9):

4882–88. http://www.ncbi.nlm.nih.gov/pubmed/12391199.

Lee, Ji-Hoon, and Tanya T Paull. 2004. “Direct Activation of the ATM Protein Kinase by the

Mre11/Rad50/Nbs1 Complex.” Science (New York, N.Y.) 304 (5667): 93–96.

doi:10.1126/science.1091496.

Lee, Youjin, Sogyong L. Auh, Yugang Wang, Byron Burnette, Yang Wang, Yuru Meng,

Michael Beckett, et al. 2009. “Therapeutic Effects of Ablative Radiation on Local Tumor

129

Require CD8 + T Cells: Changing Strategies for Cancer Treatment.” Blood 114 (3): 589–

95. doi:10.1182/blood-2009-02-206870.

Leigh, J., and Bruce Robinson. 2002. “The History of Mesothelioma in Australia 1945-2001.” In

Mesothelioma, edited by Bruce W.S. Robinson and A. Philippe Chahinian, 55–110.

London: Martin Dunitz Ltd.

Levy, Antonin, Cyrus Chargari, Morgane Cheminant, Noémie Simon, Céline Bourgier, and Eric

Deutsch. 2013. “Radiation Therapy and Immunotherapy: Implications for a Combined

Cancer Treatment.” Critical Reviews in Oncology/Hematology 85 (3). Elsevier Ireland Ltd:

278–87. doi:10.1016/j.critrevonc.2012.09.001.

Lewanski, C R, and W J Gullick. 2001. “Radiotherapy and Cellular Signalling.” The Lancet.

Oncology 2 (6): 366–70. doi:10.1016/S1470-2045(00)00391-0.

Lin, E, S E Calvano, and S F Lowry. 2000. “Inflammatory Cytokines and Cell Response in

Surgery.” Surgery 127 (2): 117–26. doi:10.1067/msy.2000.101584.

Lipson, Evan J, William H Sharfman, Charles G Drake, Ira Wollner, Janis M Taube, Robert A

Anders, Haiying Xu, et al. 2013. “Durable Cancer Regression off-Treatment and Effective

Reinduction Therapy with an Anti-PD-1 Antibody.” Clinical Cancer Research : An Official

Journal of the American Association for Cancer Research 19 (2): 462–68.

doi:10.1158/1078-0432.CCR-12-2625.

Liu, Zhiwen, and Julius Klominek. 2004. “Chemotaxis and Chemokinesis of Malignant

Mesothelioma Cells to Multiple Growth Factors.” Anticancer Research 24 (3 A): 1625–30.

Lorne, Emmanuel, Jaroslaw W Zmijewski, Xia Zhao, Gang Liu, Yuko Tsuruta, Young-Jun Park,

Hervé Dupont, and Edward Abraham. 2008. “Role of Extracellular Superoxide in

Neutrophil Activation: Interactions between Xanthine Oxidase and TLR4 Induce

Proinflammatory Cytokine Production.” American Journal of Physiology. Cell Physiology

294 (4): C985–93. doi:10.1152/ajpcell.00454.2007.

Luce, Audrey, Aurélie Courtin, Céline Levalois, Sandrine Altmeyer-Morel, Paul-Henri Romeo,

Sylvie Chevillard, and Jérôme Lebeau. 2009. “Death Receptor Pathways Mediate Targeted

130

and Non-Targeted Effects of Ionizing Radiations in Breast Cancer Cells.” Carcinogenesis

30 (3): 432–39. doi:10.1093/carcin/bgp008.

Lugade, Amit a, James P Moran, Scott a Gerber, Robert C Rose, John G Frelinger, and Edith M

Lord. 2005. “Local Radiation Therapy of B16 Melanoma Tumors Increases the Generation

of Tumor Antigen-Specific Effector Cells That Traffic to the Tumor.” Journal of

Immunology (Baltimore, Md. : 1950) 174 (12): 7516–23.

doi:10.4049/jimmunol.174.12.7516.

Lugade, Amit A, Elizabeth W Sorensen, Scott A Gerber, James P Moran, John G Frelinger, and

Edith M Lord. 2008. “Radiation-Induced IFN-Gamma Production within the Tumor

Microenvironment Influences Antitumor Immunity.” Journal of Immunology (Baltimore,

Md. : 1950) 180 (5): 3132–39. http://www.ncbi.nlm.nih.gov/pubmed/18292536.

Lumniczky, Katalin, and Géza Sáfrány. 2015. “The Impact of Radiation Therapy on the

Antitumor Immunity: Local Effects and Systemic Consequences.” Cancer Letters 356 (1):

114–25. doi:10.1016/j.canlet.2013.08.024.

Ma, Yuting, Oliver Kepp, François Ghiringhelli, Lionel Apetoh, Laetitia Aymeric, Clara Locher,

Antoine Tesniere, et al. 2010. “Chemotherapy and Radiotherapy: Cryptic Anticancer

Vaccines.” Seminars in Immunology 22 (3): 113–24. doi:10.1016/j.smim.2010.03.001.

MacConmara, Malcolm P, Adrian A Maung, Satoshi Fujimi, Ann M McKenna, Adam Delisle,

Peter H Lapchak, Selwyn Rogers, James A Lederer, and John A Mannick. 2006. “Increased

CD4+ CD25+ T Regulatory Cell Activity in Trauma Patients Depresses Protective Th1

Immunity.” Annals of Surgery 244 (4): 514–23. doi:10.1097/01.sla.0000239031.06906.1f.

Mailand, N, J Falck, C Lukas, R G Syljuâsen, M Welcker, J Bartek, and J Lukas. 2000. “Rapid

Destruction of Human Cdc25A in Response to DNA Damage.” Science (New York, N.Y.)

288 (5470): 1425–29. http://www.ncbi.nlm.nih.gov/pubmed/10827953.

Manda, Katrin, Annegret Glasow, Daniel Paape, and Guido Hildebrandt. 2012. “Effects of

Ionizing Radiation on the Immune System with Special Emphasis on the Interaction of

Dendritic and T Cells.” Frontiers in Oncology 2 (January): 102.

doi:10.3389/fonc.2012.00102.

131

Mansfield, Aaron Scott, Anja C Roden, Tobias Peikert, Yuri M Sheinin, Susan M Harrington,

Christopher J Krco, Haidong Dong, and Eugene D Kwon. 2014. “B7-H1 Expression in

Malignant Pleural Mesothelioma Is Associated with Sarcomatoid Histology and Poor

Prognosis.” Journal of Thoracic Oncology : Official Publication of the International

Association for the Study of Lung Cancer 9 (7): 1036–40.

doi:10.1097/JTO.0000000000000177.

Mantel, Frederick, Benjamin Frey, Stefan Haslinger, Petra Schildkopf, Renate Sieber, Oliver J

Ott, Barbara Lödermann, et al. 2010. “Combination of Ionising Irradiation and

Hyperthermia Activates Programmed Apoptotic and Necrotic Cell Death Pathways in

Human Colorectal Carcinoma Cells.” Strahlentherapie Und Onkologie : Organ Der

Deutschen Röntgengesellschaft ... [et Al] 186 (11): 587–99. doi:10.1007/s00066-010-2154-

x.

Marik, Paul E., and Mark Flemmer. 2012. “The Immune Response to Surgery and Trauma.”

Journal of Trauma and Acute Care Surgery 73 (4): 801–8.

doi:10.1097/TA.0b013e318265cf87.

Marinaccio, Alessandro, Fabio Montanaro, Marina Mastrantonio, Raffaella Uccelli, Pierluigi

Altavista, Massimo Nesti, Adele Seniori Costantini, and Giuseppe Gorini. 2005.

“Predictions of Mortality from Pleural Mesothelioma in Italy: A Model Based on Asbestos

Consumption Figures Supports Results from Age-Period-Cohort Models.” International

Journal of Cancer. Journal International Du Cancer 115 (1): 142–47.

doi:10.1002/ijc.20820.

Markham, A, and D Faulds. 1994. “Ganciclovir. An Update of Its Therapeutic Use in

Cytomegalovirus Infection.” Drugs 48 (3): 455–84.

http://www.ncbi.nlm.nih.gov/pubmed/7527763.

Marom, Edith M, Jeremy J Erasmus, Harvey I Pass, and Edward F Patz. 2002. “The Role of

Imaging in Malignant Pleural Mesothelioma.” Seminars in Oncology 29 (1): 26–35.

http://www.ncbi.nlm.nih.gov/pubmed/11836666.

132

Martensson, G. 1990. “Diagnosing Malignant Pleural Mesothelioma.” The European Respiratory

Journal : Official Journal of the European Society for Clinical Respiratory Physiology 3

(9): 985–86.

Masood, Rizwan, Ajay Kundra, SuTao Zhu, Guangbin Xia, Pierluigi Scalia, D Lynne Smith, and

Parkash S Gill. 2003. “Malignant Mesothelioma Growth Inhibition by Agents That Target

the VEGF and VEGF-C Autocrine Loops.” International Journal of Cancer. Journal

International Du Cancer 104 (5): 603–10. doi:10.1002/ijc.10996.

Matkowski, Rafal, Iwona Gisterek, Agnieszka Halon, Aleksandra Lacko, Krzysztof Szewczyk,

Urszula Staszek, Marek Pudelko, et al. 2009. “The Prognostic Role of Tumor-Infiltrating

CD4 and CD8 T Lymphocytes in Breast Cancer.” Anticancer Research 29 (7): 2445–51.

http://www.ncbi.nlm.nih.gov/pubmed/19596912.

Matsumura, Satoko, Baomei Wang, Noriko Kawashima, Steve Braunstein, Michelle Badura,

Thomas O Cameron, James S Babb, et al. 2008. “Radiation-Induced CXCL16 Release by

Breast Cancer Cells Attracts Effector T Cells.” Journal of Immunology (Baltimore, Md. :

1950) 181 (5): 3099–3107.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2587101&tool=pmcentrez&ren

dertype=abstract.

Matsuoka, S, G Rotman, A Ogawa, Y Shiloh, K Tamai, and S J Elledge. 2000. “Ataxia

Telangiectasia-Mutated Phosphorylates Chk2 in Vivo and in Vitro.” Proceedings of the

National Academy of Sciences of the United States of America 97 (19): 10389–94.

doi:10.1073/pnas.190030497.

Matzinger, P. 1998. “An Innate Sense of Danger.” Seminars in Immunology 10 (5): 399–415.

doi:10.1006/smim.1998.0143.

McBride, William H, Chi-Shiun Chiang, Jennifer L Olson, Chun-Chieh Wang, Ji-Hong Hong,

Frank Pajonk, Graeme J Dougherty, Keisuke S Iwamoto, Milena Pervan, and Yu-Pei Liao.

2004. “A Sense of Danger from Radiation.” Radiation Research 162 (1): 1–19.

http://www.ncbi.nlm.nih.gov/pubmed/15222781.

133

Melamed, Rivka, Shahar Bar-Yosef, Guy Shakhar, Keren Shakhar, and Shamgar Ben-Eliyahu.

2003. “Suppression of Natural Killer Cell Activity and Promotion of Tumor Metastasis by

Ketamine, Thiopental, and Halothane, but Not by Propofol: Mediating Mechanisms and

Prophylactic Measures.” Anesthesia and Analgesia 97 (5): 1331–39.

http://www.ncbi.nlm.nih.gov/pubmed/14570648.

Menger, Michael D, and Brigitte Vollmar. 2004. “Surgical Trauma: Hyperinflammation versus

Immunosuppression?” Langenbeck’s Archives of Surgery / Deutsche Gesellschaft Für

Chirurgie 389 (6): 475–84. doi:10.1007/s00423-004-0472-0.

Menges, P, W Kessler, C Kloecker, M Feuerherd, S Gaubert, S Diedrich, J van der Linde, et al.

2012. “Surgical Trauma and Postoperative Immune Dysfunction.” European Surgical

Research. Europäische Chirurgische Forschung. Recherches Chirurgicales Européennes

48 (4): 180–86. doi:10.1159/000338196.

Mezzapelle, R, U Miglio, O Rena, A Paganotti, S Allegrini, J Antona, F Molinari, et al. 2013.

“Mutation Analysis of the EGFR Gene and Downstream Signalling Pathway in Histologic

Samples of Malignant Pleural Mesothelioma.” British Journal of Cancer 108 (8): 1743–49.

doi:10.1038/bjc.2013.130.

Minatel, Emilio, Marco Trovo, Jerry Polesel, Tania Baresic, Alessandra Bearz, Giovanni

Franchin, Carlo Gobitti, et al. 2014. “Radical Pleurectomy/decortication Followed by High

Dose of Radiation Therapy for Malignant Pleural Mesothelioma. Final Results with Long-

Term Follow-Up.” Lung Cancer (Amsterdam, Netherlands) 83 (1): 78–82.

doi:10.1016/j.lungcan.2013.10.013.

Mita, Alain C, Christopher J Sweeney, Sharyn D Baker, Andrew Goetz, Lisa A Hammond,

Amita Patnaik, Anthony W Tolcher, et al. 2006. “Phase I and Pharmacokinetic Study of

Pemetrexed Administered Every 3 Weeks to Advanced Cancer Patients with Normal and

Impaired Renal Function.” Journal of Clinical Oncology : Official Journal of the American

Society of Clinical Oncology 24 (4): 552–62. doi:10.1200/JCO.2004.00.9720.

Mizuno, Kenji, Hideo Kohka Takahashi, Hiromi Iwagaki, Goutaro Katsuno, Huda A S M

Kamurul, Satoru Ohtani, Shuji Mori, Tadashi Yoshino, Masahiro Nishibori, and Noriaki

134

Tanaka. 2005. “Beta2-Adrenergic Receptor Stimulation Inhibits LPS-Induced IL-18 and IL-

12 Production in Monocytes.” Immunology Letters 101 (2): 168–72.

doi:10.1016/j.imlet.2005.05.008.

Morel, A, N Fernandez, A de La Coste, H Haddada, M Viguier, B S Polla, B Antoine, and A

Kahn. 1998. “Gamma-Ray Irradiation Induces B7.1 Costimulatory Molecule Neoexpression

in Various Murine Tumor Cells.” Cancer Immunology, Immunotherapy : CII 46 (5): 277–

82. http://www.ncbi.nlm.nih.gov/pubmed/9690456.

Morel, S, F Lévy, O Burlet-Schiltz, F Brasseur, M Probst-Kepper, A L Peitrequin, B Monsarrat,

et al. 2000. “Processing of Some Antigens by the Standard Proteasome but Not by the

Immunoproteasome Results in Poor Presentation by Dendritic Cells.” Immunity 12 (1):

107–17. http://www.ncbi.nlm.nih.gov/pubmed/10661410.

Moretti, Amanda. 2010. “Development of a Model to Study the Abscopal Effect : Combining

Image-Guided Radiation Therapy and Immunotherapy in Cancer Treatment by

Development of a Model to Study the Abscopal Effect : Combining Image-Guided

Radiation Therapy and Immunotherapy.” University of Toronto.

Mossman, Brooke T, Arti Shukla, Nicholas H Heintz, Claire F Verschraegen, Anish Thomas,

and Raffit Hassan. 2013. “New Insights into Understanding the Mechanisms, Pathogenesis,

and Management of Malignant Mesotheliomas.” The American Journal of Pathology 182

(4): 1065–77. doi:10.1016/j.ajpath.2012.12.028.

Muers, Martin F, Richard J Stephens, Patricia Fisher, Liz Darlison, Christopher M B Higgs,

Erica Lowry, Andrew G Nicholson, et al. 2008. “Active Symptom Control with or without

Chemotherapy in the Treatment of Patients with Malignant Pleural Mesothelioma (MS01):

A Multicentre Randomised Trial.” Lancet (London, England) 371 (9625): 1685–94.

doi:10.1016/S0140-6736(08)60727-8.

Nagata, S, and P Golstein. 1995. “The Fas Death Factor.” Science (New York, N.Y.) 267 (5203):

1449–56. http://www.ncbi.nlm.nih.gov/pubmed/7533326.

135

Nguyen, G K, M R Akin, R R Villanueva, and J Slatnik. 1999. “Cytopathology of Malignant

Mesothelioma of the Pleura in Fine-Needle Aspiration Biopsy.” Diagnostic Cytopathology

21 (4): 253–59. http://www.ncbi.nlm.nih.gov/pubmed/10495318.

Nishimura, T, M Nakui, M Sato, K Iwakabe, H Kitamura, M Sekimoto, A Ohta, T Koda, and S

Nishimura. 2000. “The Critical Role of Th1-Dominant Immunity in Tumor Immunology.”

Cancer Chemotherapy and Pharmacology 46 Suppl (January): S52–61.

http://www.ncbi.nlm.nih.gov/pubmed/10950149.

Norvell, Amanda. 2013. “Components of the Immune System.” In Cancer Immunotherapy, 11–

24. New Jersey: Elsevier. doi:10.1016/B978-0-12-394296-8.00002-6.

Nosho, Katsuhiko, Yoshifumi Baba, Noriko Tanaka, Kaori Shima, Marika Hayashi, Jeffrey A

Meyerhardt, Edward Giovannucci, Glenn Dranoff, Charles S Fuchs, and Shuji Ogino. 2010.

“Tumour-Infiltrating T-Cell Subsets, Molecular Changes in Colorectal Cancer, and

Prognosis: Cohort Study and Literature Review.” The Journal of Pathology 222 (4): 350–

66. doi:10.1002/path.2774.

Novy, Patricia, Michael Quigley, Xiaopei Huang, and Yiping Yang. 2007. “CD4 T Cells Are

Required for CD8 T Cell Survival during Both Primary and Memory Recall Responses.”

Journal of Immunology (Baltimore, Md. : 1950) 179 (12): 8243–51.

http://www.ncbi.nlm.nih.gov/pubmed/18056368.

Nowak, A K, M J Byrne, R Williamson, G Ryan, A Segal, D Fielding, P Mitchell, A W Musk,

and B W S Robinson. 2002. “A Multicentre Phase II Study of Cisplatin and Gemcitabine

for Malignant Mesothelioma.” British Journal of Cancer 87 (5): 491–96.

doi:10.1038/sj.bjc.6600505.

Obeid, Michel, Antoine Tesniere, François Ghiringhelli, Gian Maria Fimia, Lionel Apetoh, Jean-

Luc Perfettini, Maria Castedo, et al. 2007. “Calreticulin Exposure Dictates the

Immunogenicity of Cancer Cell Death.” Nature Medicine 13 (1): 54–61.

doi:10.1038/nm1523.

Odaka, M, D H Sterman, R Wiewrodt, Y Zhang, M Kiefer, K M Amin, G P Gao, et al. 2001.

“Eradication of Intraperitoneal and Distant Tumor by Adenovirus-Mediated Interferon-Beta

136

Gene Therapy Is Attributable to Induction of Systemic Immunity.” Cancer Research 61

(16): 6201–12. http://www.ncbi.nlm.nih.gov/pubmed/11507073.

Ogawa, K, M Hirai, T Katsube, M Murayama, K Hamaguchi, T Shimakawa, Y Naritake, T

Hosokawa, and T Kajiwara. 2000. “Suppression of Cellular Immunity by Surgical Stress.”

Surgery 127 (3): 329–36. doi:10.1067/msy.2000.103498.

Ohta, Y, V Shridhar, R K Bright, G P Kalemkerian, W Du, M Carbone, Y Watanabe, and H I

Pass. 1999. “VEGF and VEGF Type C Play an Important Role in Angiogenesis and

Lymphangiogenesis in Human Malignant Mesothelioma Tumours.” British Journal of

Cancer 81 (1): 54–61. doi:10.1038/sj.bjc.6690650.

Okuda, K, H Sasaki, O Kawano, H Yukiue, T Yokoyama, M Yano, and Y Fujii. 2008.

“Epidermal Growth Factor Receptor Gene Mutation, Amplification and Protein Expression

in Malignant Pleural Mesothelioma.” Journal of Cancer Research and Clinical Oncology

134 (10): 1105–11. doi:10.1007/s00432-008-0384-4.

Opitz, Isabelle. 2014. “Management of Malignant Pleural Mesothelioma-The European

Experience.” Journal of Thoracic Disease 6 (SUPPL.2): 238–52. doi:10.3978/j.issn.2072-

1439.2014.05.03.

Overgaard, J, and MR Horsman. 1996. “Modification of Hypoxia-Induced Radioresistance in

Tumors by the Use of Oxygen and Sensitizers.” Seminars in Radiation Oncology 6 (1): 10–

21. doi:10.1053/SRAO0060010.

Palcic, B, and L D Skarsgard. 1984. “Reduced Oxygen Enhancement Ratio at Low Doses of

Ionizing Radiation.” Radiation Research 100 (2): 328–39.

http://www.ncbi.nlm.nih.gov/pubmed/6494444.

Palucka, Karolina, and Jacques Banchereau. 2012. “Cancer Immunotherapy via Dendritic Cells.”

Nature Reviews Cancer 12 (4): 265–77. doi:10.1038/nrc3258.

Papa, Sophie, Sanjay Popat, Riyaz Shah, A Toby Prevost, Rohit Lal, Blair McLennan, Paul

Cane, et al. 2013. “Phase 2 Study of Sorafenib in Malignant Mesothelioma Previously

Treated with Platinum-Containing Chemotherapy.” Journal of Thoracic Oncology : Official

137

Publication of the International Association for the Study of Lung Cancer 8 (6): 783–87.

doi:10.1097/JTO.0b013e31828c2b26.

Park, Bonggoo, Cassian Yee, and Kyung Mi Lee. 2014. “The Effect of Radiation on the Immune

Response to Cancers.” International Journal of Molecular Sciences 15 (1): 927–43.

doi:10.3390/ijms15010927.

Park, In-Chul, Sang-Hyeok Woo, Myung-Jin Park, Hyung-Chahn Lee, Su-Jae Lee, Young-Joon

Hong, Seung-Hoon Lee, Seok-Il Hong, and Chang-Hun Rhee. “Ionizing Radiation and

Nitric Oxide Donor Sensitize Fas-Induced Apoptosis via up-Regulation of Fas in Human

Cervical Cancer Cells.” Oncology Reports 10 (3): 629–33.

http://www.ncbi.nlm.nih.gov/pubmed/12684635.

Pastan, Ira, Raffit Hassan, David J FitzGerald, and Robert J Kreitman. 2007. “Immunotoxin

Treatment of Cancer.” Annual Review of Medicine 58 (January): 221–37.

doi:10.1146/annurev.med.58.070605.115320.

Pateras, Ioannis S., Sophia Havaki, Xenia Nikitopoulou, Konstantinos Vougas, Paul Townsend,

Michalis I. Panayiotidis, Alexandros G. Georgakilas, and Vassilis G. Gorgoulis. 2015. “The

DNA Damage Response and Immune Signaling Alliance: Is It Good or Bad? Nature

Decides When and Where.” Pharmacology & Therapeutics, July.

doi:10.1016/j.pharmthera.2015.06.011.

Peng, C Y, P R Graves, R S Thoma, Z Wu, A S Shaw, and H Piwnica-Worms. 1997. “Mitotic

and G2 Checkpoint Control: Regulation of 14-3-3 Protein Binding by Phosphorylation of

Cdc25C on Serine-216.” Science (New York, N.Y.) 277 (5331): 1501–5.

http://www.ncbi.nlm.nih.gov/pubmed/9278512.

Peto, J, a Decarli, C La Vecchia, F Levi, and E Negri. 1999. “The European Mesothelioma

Epidemic.” British Journal of Cancer 79 (3-4): 666–72. doi:10.1038/sj.bjc.6690105.

Pietsch, E C, S M Sykes, S B McMahon, and M E Murphy. 2008. “The p53 Family and

Programmed Cell Death.” Oncogene 27 (50): 6507–21. doi:10.1038/onc.2008.315.

138

Pirker, Robert, Jose R Pereira, Aleksandra Szczesna, Joachim von Pawel, Maciej Krzakowski,

Rodryg Ramlau, Ihor Vynnychenko, et al. 2009. “Cetuximab plus Chemotherapy in Patients

with Advanced Non-Small-Cell Lung Cancer (FLEX): An Open-Label Randomised Phase

III Trial.” Lancet (London, England) 373 (9674): 1525–31. doi:10.1016/S0140-

6736(09)60569-9.

Pirttikangas, C O, M Salo, M Mansikka, J Grönroos, K Pulkki, and O Peltola. 1995. “The

Influence of Anaesthetic Technique upon the Immune Response to Hysterectomy. A

Comparison of Propofol Infusion and Isoflurane.” Anaesthesia 50 (12): 1056–61.

http://www.ncbi.nlm.nih.gov/pubmed/8546287.

Postow, Michael A, Margaret K Callahan, Christopher A Barker, Yoshiya Yamada, Jianda Yuan,

Shigehisa Kitano, Zhenyu Mu, et al. 2012. “Immunologic Correlates of the Abscopal Effect

in a Patient with Melanoma.” The New England Journal of Medicine 366 (10): 925–31.

doi:10.1056/NEJMoa1112824.

Postow, Michael A, James Harding, and Jedd D Wolchok. 2012. “Targeting Immune

Checkpoints: Releasing the Restraints on Anti-Tumor Immunity for Patients with

Melanoma.” Cancer Journal (Sudbury, Mass.) 18 (2): 153–59.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3751414&tool=pmcentrez&ren

dertype=abstract.

Powers, Amy, and Michele Carbone. 2002. “The Role of Environmental Carcinogens, Viruses

and Genetic Predisposition in the Pathogenesis of Mesothelioma.” Cancer Biology and

Therapy 1 (4): 348–53. doi:10.4161/cbt.1.4.4.

Puri, R K, D S Hoon, P Leland, P Snoy, R W Rand, I Pastan, and R J Kreitman. 1996.

“Preclinical Development of a Recombinant Toxin Containing Circularly Permuted

Interleukin 4 and Truncated Pseudomonas Exotoxin for Therapy of Malignant

Astrocytoma.” Cancer Research 56 (24): 5631–37.

http://www.ncbi.nlm.nih.gov/pubmed/8971168.

139

Rabinovich, Gabriel A, Dmitry Gabrilovich, and Eduardo M Sotomayor. 2007.

“Immunosuppressive Strategies That Are Mediated by Tumor Cells.” Annual Review of

Immunology 25 (January): 267–96. doi:10.1146/annurev.immunol.25.022106.141609.

Radford, I R. 1985. “The Level of Induced DNA Double-Strand Breakage Correlates with Cell

Killing after X-Irradiation.” International Journal of Radiation Biology and Related Studies

in Physics, Chemistry, and Medicine 48 (1): 45–54.

http://www.ncbi.nlm.nih.gov/pubmed/3874180.

Raja, Siva, Sudish C. Murthy, and David P. Mason. 2011. “Malignant Pleural Mesothelioma.”

Current Oncology Reports 13 (4): 259–64. doi:10.1007/s11912-011-0177-9.

Ramos-Nino, Maria E., Cynthia R. Timblin, and Brooke T. Mossman. 2002. “Mesothelial Cell

Transformation Requires Increased AP-1 Binding Activity and ERK-Dependent Fra-1

Expression.” Cancer Research 62 (21): 6065–69.

Ratzer, E R, J L Pool, and M R Melamed. 1967. “Pleural Mesotheliomas. Clinical Experiences

with Thirty-Seven Patients.” The American Journal of Roentgenology, Radium Therapy,

and Nuclear Medicine 99 (4): 863–80. http://www.ncbi.nlm.nih.gov/pubmed/6021390.

Reid, A, N de Klerk, G Ambrosini, N Olsen, S C Pang, and A W Musk. 2005. “The Additional

Risk of Malignant Mesothelioma in Former Workers and Residents of Wittenoom with

Benign Pleural Disease or Asbestosis.” Occupational and Environmental Medicine 62 (10):

665–69. doi:10.1136/oem.2004.018531.

Reits, Eric a, James W Hodge, Carla a Herberts, Tom a Groothuis, Mala Chakraborty, Elizabeth

K Wansley, Kevin Camphausen, et al. 2006. “Radiation Modulates the Peptide Repertoire,

Enhances MHC Class I Expression, and Induces Successful Antitumor Immunotherapy.”

The Journal of Experimental Medicine 203 (5): 1259–71. doi:10.1084/jem.20052494.

Richendollar, Bill G, Brad Pohlman, Paul Elson, and Eric D Hsi. 2011. “Follicular Programmed

Death 1-Positive Lymphocytes in the Tumor Microenvironment Are an Independent

Prognostic Factor in Follicular Lymphoma.” Human Pathology 42 (4): 552–57.

doi:10.1016/j.humpath.2010.08.015.

140

Robertson, John D, Vladimir Gogvadze, Andrey Kropotov, Helin Vakifahmetoglu, Boris

Zhivotovsky, and Sten Orrenius. 2004. “Processed Caspase-2 Can Induce Mitochondria-

Mediated Apoptosis Independently of Its Enzymatic Activity.” EMBO Reports 5 (6): 643–

48. doi:10.1038/sj.embor.7400153.

Robinson, Benjamin M. 2012. “Malignant Pleural Mesothelioma: An Epidemiological

Perspective.” Annals of Cardiothoracic Surgery 1 (4): 491–96. doi:10.3978/j.issn.2225-

319X.2012.11.04.

Robinson, Bruce W S, and Richard A Lake. 2005. “Advances in Malignant Mesothelioma.” The

New England Journal of Medicine 353 (15): 1591–1603. doi:10.1056/NEJMra050152.

Robinson, Bruce W S, Arthur W Musk, and Richard A Lake. 2005. “Malignant Mesothelioma.”

Lancet (London, England) 366 (9483): 397–408. doi:10.1016/S0140-6736(05)67025-0.

Rock, Kenneth L, and Hajime Kono. 2008. “The Inflammatory Response to Cell Death.” Annual

Review of Pathology 3 (January): 99–126.

doi:10.1146/annurev.pathmechdis.3.121806.151456.

Roedel, F, N Kley, H U Beuscher, G Hildebrandt, L Keilholz, P Kern, R Voll, M Herrmann, and

R Sauer. 2002. “Anti-Inflammatory Effect of Low-Dose X-Irradiation and the Involvement

of a TGF-beta1-Induced down-Regulation of Leukocyte/endothelial Cell Adhesion.”

International Journal of Radiation Biology 78 (8): 711–19.

doi:10.1080/09553000210137671.

Rosenne, Ella, Liat Sorski, Lee Shaashua, Elad Neeman, Pini Matzner, Ben Levi, and Shamgar

Ben-Eliyahu. 2014. “In Vivo Suppression of NK Cell Cytotoxicity by Stress and Surgery:

Glucocorticoids Have a Minor Role Compared to Catecholamines and Prostaglandins.”

Brain, Behavior, and Immunity 37 (March): 207–19. doi:10.1016/j.bbi.2013.12.007.

Rosenzweig, Kenneth E, Marjorie G Zauderer, Benjamin Laser, Lee M Krug, Ellen Yorke,

Camelia S Sima, Andreas Rimner, Raja Flores, and Valerie Rusch. 2012. “Pleural Intensity-

Modulated Radiotherapy for Malignant Pleural Mesothelioma.” International Journal of

Radiation Oncology, Biology, Physics 83 (4): 1278–83. doi:10.1016/j.ijrobp.2011.09.027.

141

Rovere-Querini, Patrizia, Annalisa Capobianco, Paola Scaffidi, Barbara Valentinis, Federica

Catalanotti, Marta Giazzon, Ingrid E Dumitriu, et al. 2004. “HMGB1 Is an Endogenous

Immune Adjuvant Released by Necrotic Cells.” EMBO Reports 5 (8): 825–30.

doi:10.1038/sj.embor.7400205.

Rübe, Claudia E, Falk Wilfert, Jan Palm, Jochem König, Susanne Burdak-Rothkamm, Li Liu,

Andreas Schuck, Normann Willich, and Christian Rübe. 2004. “Irradiation Induces a

Biphasic Expression of pro-Inflammatory Cytokines in the Lung.” Strahlentherapie Und

Onkologie : Organ Der Deutschen Röntgengesellschaft ... [et Al] 180 (7): 442–48.

doi:10.1007/s00066-004-1265-7.

Ruffie, P, R Feld, S Minkin, Y Cormier, A Boutan-Laroze, R Ginsberg, J Ayoub, F A Shepherd,

W K Evans, and A Figueredo. 1989. “Diffuse Malignant Mesothelioma of the Pleura in

Ontario and Quebec: A Retrospective Study of 332 Patients.” Journal of Clinical

Oncology : Official Journal of the American Society of Clinical Oncology 7 (8): 1157–68.

http://www.ncbi.nlm.nih.gov/pubmed/2666592.

Rusch, V W, K Rosenzweig, E Venkatraman, L Leon, A Raben, L Harrison, M S Bains, R J

Downey, and R J Ginsberg. 2001. “A Phase II Trial of Surgical Resection and Adjuvant

High-Dose Hemithoracic Radiation for Malignant Pleural Mesothelioma.” The Journal of

Thoracic and Cardiovascular Surgery 122 (4): 788–95. doi:10.1067/mtc.2001.116560.

Rusch, Valerie, Elizabeth H. Baldini, Raphael Bueno, Marc De Perrot, Raja Flores, Seiki

Hasegawa, Walter Klepetko, et al. 2013. “The Role of Surgical Cytoreduction in the

Treatment of Malignant Pleural Mesothelioma: Meeting Summary of the International

Mesothelioma Interest Group Congress, September 11-14, 2012, Boston, Mass.” Journal of

Thoracic and Cardiovascular Surgery 145 (4): 909–10. doi:10.1016/j.jtcvs.2013.01.039.

Rusch, Valerie W., Dorothy Giroux, Catherine Kennedy, Enrico Ruffini, Ayten K. Cangir, David

Rice, Harvey Pass, et al. 2012. “Initial Analysis of the International Association For the

Study of Lung Cancer Mesothelioma Database.” Journal of Thoracic Oncology 7 (11):

1631–39. doi:10.1097/JTO.0b013e31826915f1.

142

Russell, John H, and Timothy J Ley. 2002. “Lymphocyte-Mediated Cytotoxicity.” Annual

Review of Immunology 20 (January): 323–70.

doi:10.1146/annurev.immunol.20.100201.131730.

Sanchez, Y, C Wong, R S Thoma, R Richman, Z Wu, H Piwnica-Worms, and S J Elledge. 1997.

“Conservation of the Chk1 Checkpoint Pathway in Mammals: Linkage of DNA Damage to

Cdk Regulation through Cdc25.” Science (New York, N.Y.) 277 (5331): 1497–1501.

http://www.ncbi.nlm.nih.gov/pubmed/9278511.

Sanders, C L, and T A Jackson. 1972. “Induction of Mesotheliomas and Sarcomas from ‘Hot

Spots’ of 239 PuO 2 Activity.” Health Physics 22 (6): 755–59.

http://www.ncbi.nlm.nih.gov/pubmed/5084007.

Sanford, Mark. 2012. “Ipilimumab: In Previously Treated Patients with Advanced Melanoma.”

BioDrugs : Clinical Immunotherapeutics, Biopharmaceuticals and Gene Therapy 26 (3):

185–93. doi:10.2165/11208200-000000000-00000.

Saoulli, K, S Y Lee, J L Cannons, W C Yeh, A Santana, M D Goldstein, N Bangia, et al. 1998.

“CD28-Independent, TRAF2-Dependent Costimulation of Resting T Cells by 4-1BB

Ligand.” The Journal of Experimental Medicine 187 (11): 1849–62.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2212301&tool=pmcentrez&ren

dertype=abstract.

Seki, Naoko, Alan D Brooks, Clive R D Carter, Timothy C Back, Erin M Parsoneault, Mark J

Smyth, Robert H Wiltrout, and Thomas J Sayers. 2002. “Tumor-Specific CTL Kill Murine

Renal Cancer Cells Using Both Perforin and Fas Ligand-Mediated Lysis in Vitro, but Cause

Tumor Regression in Vivo in the Absence of Perforin.” Journal of Immunology (Baltimore,

Md. : 1950) 168 (7): 3484–92. http://www.ncbi.nlm.nih.gov/pubmed/11907109.

Seo, A, F Ishikawa, H Nakano, H Nakazaki, K Kobayashi, and T Kakiuchi. 1999. “Enhancement

of B7-1 (CD80) Expression on B-Lymphoma Cells by Irradiation.” Immunology 96 (4):

642–48.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2326785&tool=pmcentrez&ren

dertype=abstract.

143

Sgadari, C, A L Angiolillo, and G Tosato. 1996. “Inhibition of Angiogenesis by Interleukin-12 Is

Mediated by the Interferon-Inducible Protein 10.” Blood 87 (9): 3877–82.

http://www.ncbi.nlm.nih.gov/pubmed/8611715.

Shankaran, V, H Ikeda, A T Bruce, J M White, P E Swanson, L J Old, and R D Schreiber. 2001.

“IFNgamma and Lymphocytes Prevent Primary Tumour Development and Shape Tumour

Immunogenicity.” Nature 410 (6832): 1107–11. doi:10.1038/35074122.

Sheard, Michael A, Stjepan Uldrijan, and Borivoj Vojtesek. 2003. “Role of p53 in Regulating

Constitutive and X-Radiation-Inducible CD95 Expression and Function in Carcinoma

Cells.” Cancer Research 63 (21): 7176–84.

http://www.ncbi.nlm.nih.gov/pubmed/14612511.

Shieh, S Y, J Ahn, K Tamai, Y Taya, and C Prives. 2000. “The Human Homologs of Checkpoint

Kinases Chk1 and Cds1 (Chk2) Phosphorylate p53 at Multiple DNA Damage-Inducible

Sites.” Genes & Development 14 (3): 289–300.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=316358&tool=pmcentrez&rend

ertype=abstract.

Shiraishi, Kenshiro, Yoshiro Ishiwata, Keiichi Nakagawa, Shoji Yokochi, Chiho Taruki, Teruo

Akuta, Kuni Ohtomo, Kouji Matsushima, Takuya Tamatani, and Shiro Kanegasaki. 2008.

“Enhancement of Antitumor Radiation Efficacy and Consistent Induction of the Abscopal

Effect in Mice by ECI301, an Active Variant of Macrophage Inflammatory Protein-1alpha.”

Clinical Cancer Research : An Official Journal of the American Association for Cancer

Research 14 (4): 1159–66. doi:10.1158/1078-0432.CCR-07-4485.

Shivapurkar, Narayan, Kenichi Harada, Jyotsna Reddy, Richard H Scheuermann, Yin Xu, Robert

W McKenna, Sara Milchgrub, Steven H Kroft, Ziding Feng, and Adi F Gazdar. 2002.

“Presence of Simian Virus 40 DNA Sequences in Human Lymphomas.” Lancet 359 (9309):

851–52. doi:10.1016/S0140-6736(02)07921-7.

Silva, Manuel T. 2010. “Secondary Necrosis: The Natural Outcome of the Complete Apoptotic

Program.” FEBS Letters 584 (22): 4491–99. doi:10.1016/j.febslet.2010.10.046.

144

Son, Cheol-Hun, Jae-Ho Bae, Dong-Yeok Shin, Hong-Rae Lee, Wol-Soon Jo, Kwangmo Yang,

and You-Soo Park. 2015. “Combination Effect of Regulatory T-Cell Depletion and Ionizing

Radiation in Mouse Models of Lung and Colon Cancer.” International Journal of Radiation

Oncology, Biology, Physics 92 (2): 390–98. doi:10.1016/j.ijrobp.2015.01.011.

Soussi, T, and G Lozano. 2005. “p53 Mutation Heterogeneity in Cancer.” Biochemical and

Biophysical Research Communications 331 (3): 834–42. doi:10.1016/j.bbrc.2005.03.190.

Stahel, R. a., W. Weder, Y. Lievens, and E. Felip. 2010. “Malignant Pleural Mesothelioma:

ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up.” Annals of

Oncology 21 (SUPPL. 5): 2009–11. doi:10.1093/annonc/mdq173.

Stebbing, Justin, Thomas Powles, Kirsty McPherson, Jonathan Shamash, Paula Wells, Michael T

Sheaff, Sarah Slater, Robin M Rudd, Dean Fennell, and Jeremy P C Steele. 2009. “The

Efficacy and Safety of Weekly Vinorelbine in Relapsed Malignant Pleural Mesothelioma.”

Lung Cancer (Amsterdam, Netherlands) 63 (1): 94–97. doi:10.1016/j.lungcan.2008.04.001.

Sterman, Daniel H, Andrew Haas, Edmund Moon, Adriana Recio, Daniel Schwed, Anil Vachani,

Sharyn I Katz, et al. 2011. “A Trial of Intrapleural Adenoviral-Mediated Interferon-α2b

Gene Transfer for Malignant Pleural Mesothelioma.” American Journal of Respiratory and

Critical Care Medicine 184 (12): 1395–99. doi:10.1164/rccm.201103-0554CR.

Sterman, Daniel H, Adri Recio, Richard G Carroll, Colin T Gillespie, Andrew Haas, Anil

Vachani, Veena Kapoor, et al. 2007. “A Phase I Clinical Trial of Single-Dose Intrapleural

IFN-Beta Gene Transfer for Malignant Pleural Mesothelioma and Metastatic Pleural

Effusions: High Rate of Antitumor Immune Responses.” Clinical Cancer Research : An

Official Journal of the American Association for Cancer Research 13 (15 Pt 1): 4456–66.

doi:10.1158/1078-0432.CCR-07-0403.

Sterman, Daniel H, Adriana Recio, Anil Vachani, Jing Sun, Lumei Cheung, Peter DeLong,

Kunjlata M Amin, et al. 2005. “Long-Term Follow-up of Patients with Malignant Pleural

Mesothelioma Receiving High-Dose Adenovirus Herpes Simplex Thymidine

Kinase/ganciclovir Suicide Gene Therapy.” Clinical Cancer Research : An Official Journal

145

of the American Association for Cancer Research 11 (20): 7444–53. doi:10.1158/1078-

0432.CCR-05-0405.

Strickler, H D, P S Rosenberg, S S Devesa, J Hertel, J F Fraumeni, and J J Goedert. 1998.

“Contamination of Poliovirus Vaccines with Simian Virus 40 (1955-1963) and Subsequent

Cancer Rates.” JAMA 279 (4): 292–95. http://www.ncbi.nlm.nih.gov/pubmed/9450713.

Stutman. 1975. “Immunodepression and Malignancy.” Advances in Cancer Research 22

(January): 261–422. http://www.ncbi.nlm.nih.gov/pubmed/766581.

Stutman, O. 1974. “Tumor Development after 3-Methylcholanthrene in Immunologically

Deficient Athymic-Nude Mice.” Science (New York, N.Y.) 183 (4124): 534–36.

http://www.ncbi.nlm.nih.gov/pubmed/4588620.

Stutman, Osias. 1979. “Chemical Carcinogenesis in Nude Mice: Comparison between Nude

Mice from Homozygous Matings and Heterozygous Matings and Effect of Age and

Carcinogen Dose.” Journal of the National Cancer Institute 62 (2): 353–58.

http://www.ncbi.nlm.nih.gov/pubmed/283266.

Sun, Joseph C, Matthew A Williams, and Michael J Bevan. 2004. “CD4+ T Cells Are Required

for the Maintenance, Not Programming, of Memory CD8+ T Cells after Acute Infection.”

Nature Immunology 5 (9): 927–33. doi:10.1038/ni1105.

Suzuki, Yasunosuke, Steven R Yuen, and Richard Ashley. 2005. “Short, Thin Asbestos Fibers

Contribute to the Development of Human Malignant Mesothelioma: Pathological

Evidence.” International Journal of Hygiene and Environmental Health 208 (3): 201–10.

doi:10.1016/j.ijheh.2005.01.015.

Tagliabue, Elda, Roberto Agresti, Maria Luisa Carcangiu, Cristina Ghirelli, Daniele Morelli,

Manuela Campiglio, Maritza Martel, et al. 2003. “Role of HER2 in Wound-Induced Breast

Carcinoma Proliferation.” Lancet (London, England) 362 (9383): 527–33.

doi:10.1016/S0140-6736(03)14112-8.

Takeshima, Tsuguhide, Kenji Chamoto, Daiko Wakita, Takayuki Ohkuri, Yuji Togashi, Hiroki

Shirato, Hidemitsu Kitamura, and Takashi Nishimura. 2010. “Local Radiation Therapy

146

Inhibits Tumor Growth through the Generation of Tumor-Specific CTL: Its Potentiation by

Combination with TH1 Cell Therapy.” Cancer Research 70 (7): 2697–2706.

doi:10.1158/0008-5472.CAN-09-2982.

Tan, Y, M Xu, W Wang, F Zhang, D Li, X Xu, J Gu, and R M Hoffman. “IL-2 Gene Therapy of

Advanced Lung Cancer Patients.” Anticancer Research 16 (4A): 1993–98.

http://www.ncbi.nlm.nih.gov/pubmed/8712732.

Tanaka, S, N Choe, a Iwagaki, D R Hemenway, and E Kagan. 2000. “Asbestos Exposure

Induces MCP-1 Secretion by Pleural Mesothelial Cells.” Experimental Lung Research 26

(4): 241–55.

Tang, P, S K Vatsia, S Teichberg, and E Kahn. 2001. “Pulmonary Adenocarcinoma Simulating

Malignant Mesothelioma.” Archives of Pathology & Laboratory Medicine 125 (12): 1598–

1600. doi:10.1043/0003-9985(2001)125<1598:PASMM>2.0.CO;2.

Tanida, Satoshi, Tsutomu Mizoshita, Keiji Ozeki, Hironobu Tsukamoto, Takeshi Kamiya,

Hiromi Kataoka, Daitoku Sakamuro, and Takashi Joh. 2012. “Mechanisms of Cisplatin-

Induced Apoptosis and of Cisplatin Sensitivity: Potential of BIN1 to Act as a Potent

Predictor of Cisplatin Sensitivity in Gastric Cancer Treatment.” International Journal of

Surgical Oncology 2012 (January): 862879. doi:10.1155/2012/862879.

Tesniere, A, T Panaretakis, O Kepp, L Apetoh, F Ghiringhelli, L Zitvogel, and G Kroemer. 2008.

“Molecular Characteristics of Immunogenic Cancer Cell Death.” Cell Death and

Differentiation 15 (1): 3–12. doi:10.1038/sj.cdd.4402269.

Thompson, R Houston, Haidong Dong, Christine M Lohse, Bradley C Leibovich, Michael L

Blute, John C Cheville, and Eugene D Kwon. 2007. “PD-1 Is Expressed by Tumor-

Infiltrating Immune Cells and Is Associated with Poor Outcome for Patients with Renal Cell

Carcinoma.” Clinical Cancer Research : An Official Journal of the American Association

for Cancer Research 13 (6): 1757–61. doi:10.1158/1078-0432.CCR-06-2599.

Till, Brian G, Michael C Jensen, Jinjuan Wang, Eric Y Chen, Brent L Wood, Harvey A

Greisman, Xiaojun Qian, et al. 2008. “Adoptive Immunotherapy for Indolent Non-Hodgkin

147

Lymphoma and Mantle Cell Lymphoma Using Genetically Modified Autologous CD20-

Specific T Cells.” Blood 112 (6): 2261–71. doi:10.1182/blood-2007-12-128843.

Till, Brian G, Michael C Jensen, Jinjuan Wang, Xiaojun Qian, Ajay K Gopal, David G Maloney,

Catherine G Lindgren, et al. 2012. “CD20-Specific Adoptive Immunotherapy for

Lymphoma Using a Chimeric Antigen Receptor with Both CD28 and 4-1BB Domains: Pilot

Clinical Trial Results.” Blood 119 (17): 3940–50. doi:10.1182/blood-2011-10-387969.

Tischoff, Iris, Matthias Neid, Volker Neumann, and Andrea Tannapfel. 2011. “Pathohistological

Diagnosis and Differential Diagnosis.” Recent Results in Cancer Research. Fortschritte Der

Krebsforschung. Progrès Dans Les Recherches Sur Le Cancer 189 (January): 57–78.

doi:10.1007/978-3-642-10862-4_5.

Topalian, Suzanne L, Charles G Drake, and Drew M Pardoll. 2012. “Targeting the PD-1/B7-

H1(PD-L1) Pathway to Activate Anti-Tumor Immunity.” Current Opinion in Immunology

24 (2): 207–12. doi:10.1016/j.coi.2011.12.009.

Topalian, Suzanne L, F Stephen Hodi, Julie R Brahmer, Scott N Gettinger, David C Smith,

David F McDermott, John D Powderly, et al. 2012. “Safety, Activity, and Immune

Correlates of Anti-PD-1 Antibody in Cancer.” The New England Journal of Medicine 366

(26): 2443–54. doi:10.1056/NEJMoa1200690.

Travis, Lois B, Sophie D Fosså, Sara J Schonfeld, Mary L McMaster, Charles F Lynch, Hans

Storm, Per Hall, et al. 2005. “Second Cancers among 40,576 Testicular Cancer Patients:

Focus on Long-Term Survivors.” Journal of the National Cancer Institute 97 (18): 1354–

65. doi:10.1093/jnci/dji278.

Treasure, Tom, Loic Lang-Lazdunski, David Waller, Judith M. Bliss, Carol Tan, James Entwisle,

Michael Snee, et al. 2011. “Extra-Pleural Pneumonectomy versus No Extra-Pleural

Pneumonectomy for Patients with Malignant Pleural Mesothelioma: Clinical Outcomes of

the Mesothelioma and Radical Surgery (MARS) Randomised Feasibility Study.” The

Lancet Oncology 12 (8). Elsevier Ltd: 763–72. doi:10.1016/S1470-2045(11)70149-8.

148

Tsuchiya, Yasunori, Shigeaki Sawada, Isaku Yoshioka, Yasukata Ohashi, Mitsuhiro Matsuo,

Yuko Harimaya, Kazuhiro Tsukada, and Ikuo Saiki. 2003. “Increased Surgical Stress

Promotes Tumor Metastasis.” Surgery 133 (5): 547–55. doi:10.1067/msy.2003.141.

Vachani, Anil, Edmund Moon, Elliot Wakeam, and Steven M. Albelda. 2010. “Gene Therapy for

Mesothelioma and Lung Cancer.” American Journal of Respiratory Cell and Molecular

Biology 42 (4): 385–93. doi:10.1165/rcmb.2010-0026RT.

Valerie, Kristoffer, and Lawrence F Povirk. 2003. “Regulation and Mechanisms of Mammalian

Double-Strand Break Repair.” Oncogene 22 (37): 5792–5812. doi:10.1038/sj.onc.1206679.

Van den Broek, M E, D Kägi, F Ossendorp, R Toes, S Vamvakas, W K Lutz, C J Melief, R M

Zinkernagel, and H Hengartner. 1996. “Decreased Tumor Surveillance in Perforin-Deficient

Mice.” The Journal of Experimental Medicine 184 (5): 1781–90.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2192859&tool=pmcentrez&ren

dertype=abstract.

Van Valen, F, V Kentrup-Lardong, B Truckenbrod, C Rübe, W Winkelmann, and W W Jürgens.

1997. “Regulation of the Release of Tumour Necrosis Factor (TNF)alpha and Soluble TNF

Receptor by Gamma Irradiation and Interferon Gamma in Ewing’s Sarcoma/peripheral

Primitive Neuroectodermal Tumour Cells.” Journal of Cancer Research and Clinical

Oncology 123 (5): 245–52. http://www.ncbi.nlm.nih.gov/pubmed/9201246.

Vandenabeele, Peter, Lorenzo Galluzzi, Tom Vanden Berghe, and Guido Kroemer. 2010.

“Molecular Mechanisms of Necroptosis: An Ordered Cellular Explosion.” Nature Reviews.

Molecular Cell Biology 11 (10): 700–714. doi:10.1038/nrm2970.

Vanpouille-Box, Claire, Karsten A Pilones, Erik Wennerberg, Silvia C Formenti, and Sandra

Demaria. 2015. “In Situ Vaccination by Radiotherapy to Improve Responses to Anti-

CTLA-4 Treatment.” Vaccine, July. doi:10.1016/j.vaccine.2015.05.105.

Victor, Christina Twyman-Saint, Andrew J. Rech, Amit Maity, Ramesh Rengan, Kristen E.

Pauken, Erietta Stelekati, Joseph L. Benci, et al. 2015. “Radiation and Dual Checkpoint

Blockade Activate Non-Redundant Immune Mechanisms in Cancer.” Nature 520 (7547):

373–77. doi:10.1038/nature14292.

149

Vogelzang, Nicholas J., James J. Rusthoven, James Symanowski, Claude Denham, E. Kaukel,

Pierre Ruffie, Ulrich Gatzemeier, et al. 2003. “Phase III Study of Pemetrexed in

Combination with Cisplatin versus Cisplatin Alone in Patients with Malignant Pleural

Mesothelioma.” Journal of Clinical Oncology 21 (14): 2636–44.

doi:10.1200/JCO.2003.11.136.

Voll, R E, M Herrmann, E A Roth, C Stach, J R Kalden, and I Girkontaite. 1997.

“Immunosuppressive Effects of Apoptotic Cells.” Nature 390 (6658): 350–51.

doi:10.1038/37022.

Wada, Hiroki, Shuhji Seki, Tetsuya Takahashi, Nobuaki Kawarabayashi, Hideyuki Higuchi,

Yoshiko Habu, Shinya Sugahara, and Tomiei Kazama. 2007. “Combined Spinal and

General Anesthesia Attenuates Liver Metastasis by Preserving TH1/TH2 Cytokine

Balance.” Anesthesiology 106 (3): 499–506.

http://www.ncbi.nlm.nih.gov/pubmed/17325508.

Wagner, J C, C a Sleggs, and P Marchand. 1960. “Diffuse Pleural Mesothelioma and Asbestos

Exposure in the North Western Cape Province.” British Journal of Industrial Medicine 17:

260–71.

Wang, Yaohe, Steven P Faux, Gunnel Hallden, David H Kirn, Cathy E Houghton, Nicholas R

Lemoine, and Graham Patrick. 2004. “Interleukin-1beta and Tumour Necrosis Factor-Alpha

Promote the Transformation of Human Immortalised Mesothelial Cells by Erionite.”

International Journal of Oncology 25 (1): 173–78.

http://www.ncbi.nlm.nih.gov/pubmed/15202003.

Weber, J S, and S A Rosenberg. 1988. “Modulation of Murine Tumor Major Histocompatibility

Antigens by Cytokines in Vivo and in Vitro.” Cancer Research 48 (20): 5818–24.

http://www.ncbi.nlm.nih.gov/pubmed/3139284.

Welden, Brett, Gerald Gates, Resti Mallari, and Normalynn Garrett. 2009. “Effects of

Anesthetics and Analgesics on Natural Killer Cell Activity.” AANA Journal 77 (4): 287–92.

http://www.ncbi.nlm.nih.gov/pubmed/19731847.

150

Whitaker, D. 2000. “The Cytology of Malignant Mesothelioma.” Cytopathology : Official

Journal of the British Society for Clinical Cytology 11 (3): 139–51.

http://www.ncbi.nlm.nih.gov/pubmed/10877273.

Withers, HR. 1993. “Treatment-Induced Accelerated Human Tumor Growth.” Seminars in

Radiation Oncology 3 (2): 135–43. doi:10.1054/SRAO00300135.

Wu, L., Z. Yun, T. Tagawa, K. Rey-McIntyre, and M. de Perrot. 2012. “CTLA-4 Blockade

Expands Infiltrating T Cells and Inhibits Cancer Cell Repopulation during the Intervals of

Chemotherapy in Murine Mesothelioma.” Molecular Cancer Therapeutics 11 (8): 1809–19.

doi:10.1158/1535-7163.MCT-11-1014.

Wu, Licun, Matthew Onn Wu, Luis De Maza, Zhihong Yun, Julie Yu, John Cho, and Marc De

Perrot. 2015. “Targeting the Inhibitory Receptor CTLA-4 on T Cells Increased Apscopal

Effects in Murine Mesothelioma Model.”

Wu, Licun, Zhihong Yun, Tetsuzo Tagawa, Katrina Rey-McIntyre, Masaki Anraku, and Marc de

Perrot. 2011. “Tumor Cell Repopulation between Cycles of Chemotherapy Is Inhibited by

Regulatory T-Cell Depletion in a Murine Mesothelioma Model.” Journal of Thoracic

Oncology : Official Publication of the International Association for the Study of Lung

Cancer 6 (9): 1578–86. doi:10.1097/JTO.0b013e3182208ee0.

Xie, Q W, R Whisnant, and C Nathan. 1993. “Promoter of the Mouse Gene Encoding Calcium-

Independent Nitric Oxide Synthase Confers Inducibility by Interferon Gamma and Bacterial

Lipopolysaccharide.” The Journal of Experimental Medicine 177 (6): 1779–84.

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2191051&tool=pmcentrez&ren

dertype=abstract.

Xing, Z, J Gauldie, G Cox, H Baumann, M Jordana, X F Lei, and M K Achong. 1998. “IL-6 Is

an Antiinflammatory Cytokine Required for Controlling Local or Systemic Acute

Inflammatory Responses.” Journal of Clinical Investigation 101 (2): 311–20.

doi:10.1172/JCI1368.

Xu, An, Hongning Zhou, Dennis Zengliang Yu, and Tom K. Hei. 2002. “Mechanisms of the

Genotoxicity of Crocidolite Asbestos in Mammalian Cells: Implication from Mutation

151

Patterns Induced by Reactive Oxygen Species.” Environmental Health Perspectives 110

(10): 1003–8. doi:10.1289/ehp.021101003.

Yang, Haining, Maurizio Bocchetta, Barbara Kroczynska, Amira G Elmishad, Yuanbin Chen,

Zemin Liu, Concetta Bubici, et al. 2006. “TNF-Alpha Inhibits Asbestos-Induced

Cytotoxicity via a NF-kappaB-Dependent Pathway, a Possible Mechanism for Asbestos-

Induced Oncogenesis.” Proceedings of the National Academy of Sciences of the United

States of America 103 (27): 10397–402. doi:10.1073/pnas.0604008103.

Yang, Haining, Joseph R. Testa, and Michele Carbone. 2008. “Mesothelioma Epidemiology,

Carcinogenesis, and Pathogenesis.” Current Treatment Options in Oncology 9 (2-3): 147–

57. doi:10.1007/s11864-008-0067-z.

Yarborough, Charles M. 2006. “Chrysotile as a Cause of Mesothelioma: An Assessment Based

on Epidemiology.” Critical Reviews in Toxicology 36 (2): 165–87.

http://www.ncbi.nlm.nih.gov/pubmed/16736942.

Yom, Sue S. 2015. “Accelerated Repopulation as a Cause of Radiation Treatment Failure in

Non-Small Cell Lung Cancer: Review of Current Data and Future Clinical Strategies.”

Seminars in Radiation Oncology 25 (2): 93–99. doi:10.1016/j.semradonc.2014.12.002.

Zalcman, Gerard, Julien Mazieres, Jacques Margery, Laurent Greillier, Clarisse Audigier-

Valette, Denis Moro-Sibilot, Oliver Molinier, et al. 2015. “Bevacizumab 15mg/kg plus

Cisplatin-Pemetrexed (CP) Triplet versus CP Doublet in Malignant Pleural Mesothelioma

(MPM): Results of the IFCT-GFPC-0701 MAPS Randomized Phase 3 Trial.” Journal of

Clinical Investigations (ASCO Meeting Abstracts) 33 (15_suppl).

http://hwmaint.meeting.ascopubs.org/cgi/content/abstract/33/15_suppl/7500.

Zhang, Bin, Natalie A Bowerman, Joseph K Salama, Hank Schmidt, Michael T Spiotto, Andrea

Schietinger, Ping Yu, et al. 2007. “Induced Sensitization of Tumor Stroma Leads to

Eradication of Established Cancer by T Cells.” The Journal of Experimental Medicine 204

(1): 49–55. doi:10.1084/jem.20062056.

Zhang, Y, T C Lee, B Guillemin, M C Yu, and W N Rom. 1993. “Enhanced IL-1 Beta and

Tumor Necrosis Factor-Alpha Release and Messenger RNA Expression in Macrophages

152

from Idiopathic Pulmonary Fibrosis or after Asbestos Exposure.” Journal of Immunology

(Baltimore, Md. : 1950) 150 (9): 4188–96. http://www.ncbi.nlm.nih.gov/pubmed/8473757.

Zhao, Yangbing, Edmund Moon, Carmine Carpenito, Chrystal M Paulos, Xiaojun Liu, Andrea L

Brennan, Anne Chew, et al. 2010. “Multiple Injections of Electroporated Autologous T

Cells Expressing a Chimeric Antigen Receptor Mediate Regression of Human Disseminated

Tumor.” Cancer Research 70 (22): 9053–61. doi:10.1158/0008-5472.CAN-10-2880.

Zhu, Xinmei, Gabriel Herrera, and Juan B Ochoa. 2010. “Immunosupression and Infection after

Major Surgery: A Nutritional Deficiency.” Critical Care Clinics 26 (3): 491–500, ix.

doi:10.1016/j.ccc.2010.04.004.

Zou, Weiping, and Lieping Chen. 2008. “Inhibitory B7-Family Molecules in the Tumour

Microenvironment.” Nature Reviews. Immunology 8 (6): 467–77. doi:10.1038/nri2326.