02.12.2016 Views

Journal Thoracic Oncology

WCLC2016-Abstract-Book_vF-WEB_revNov17-1

WCLC2016-Abstract-Book_vF-WEB_revNov17-1

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Abstracts <strong>Journal</strong> of <strong>Thoracic</strong> <strong>Oncology</strong> • Volume 12 Issue S1 January 2017<br />

MESOTHELIOMA<br />

Isabelle Opitz<br />

Division of <strong>Thoracic</strong> Surgery, University Hospital Zurich, Zurich/Switzerland<br />

The rational of localized / intracavitary treatment is to eliminate microscopic<br />

residual disease (MRD) after macroscopic complete resection (MCR) for<br />

mesothelioma patients. The advantage of the treatment is that local effects<br />

can be enhanced whereas systemic side effects of the therapeutic agents<br />

applied might be reduced. Several approaches have been investigated<br />

over the past decades in preclinical and clinical trials, such as intracavitary<br />

chemotherapy (iCTX), immunotherapy (iIT), photodynamic therapy (PDT)<br />

and gene therapy (iGT). Intracavitary chemotherapy (iCTX) iCTX has<br />

been studied after mesothelioma resection, not only after extrapleural<br />

pneumonectomy (EPP) but also after (extended) pleurectomy/decortication<br />

((e)P/D). The main therapeutic agent used is cisplatin. In some trials<br />

hyperthermia was additionally added with the aims to enhance the<br />

penetration of cisplatin into tissues and maximize its cytotoxicity in tumor<br />

cells [1]. Hyperthermic intrapleural perfusion had a maximum tolerated<br />

dose of 225-250 mg Cisplatin/m 2 BSA [2]. The morbidity ranges from 13 to<br />

85% and the mortality from 0 to 29% [2, 3]. Some complications are related<br />

to renal toxicity which was the dose limiting adverse event [2]. Median<br />

survival time reaches up to 35.3 months in low-risk MPM patients receiving<br />

hyperthermic intraplueral cisplatin chemotherapy following MCR [4]. This<br />

treatment is only considered for well-designed clinical trials. In vivo preclinical<br />

model using intrapleural administration of cisplatin-mixed loaded to a fibrin<br />

carrier improved the local drug concentration and prolonged the exposure<br />

of tissue to high cisplatin concentration [5]. Our phase I dose escalation<br />

trial (INFLuenCe – Meso; see figure) has proven the safety of this treatment<br />

approach (manuscript in preparation). This treatment regimen is now being<br />

tested in a phase II trial (NCT01644994).<br />

In addition to chemotherapy, other substances have also been tested for<br />

intracavitary treatment. Recently, Tada, et. al. reported study plan for a phase<br />

I trial for intrapleural treatment with zoledronic acid, a third generation of<br />

bisphosphonates, in inoperable MPM, after having successfully proven the<br />

efficacy of zoledronic in a pre-clinical model [6]. Intracavitary<br />

immunotherapy (iIT) MPM is not a classical “immunogenic” tumor.<br />

Intrapleural instillation of cytokines such as interleukin (IL)-2, interferon<br />

(IFN)-α and IFN-γ provided a good control of malignant pleural effusion (MPE)<br />

and MPM with minimal toxicity [7, 8]. To prolong and increase local exposure<br />

of IFNs, recent studies implemented immuno-gene therapy approach using<br />

adenoviral vector expressing human IFNs. Four out of 10 patients with MPM<br />

and metastatic pleural effusions showed stable disease following a single<br />

dose of intrapleural adenoviral vector expressing IFN-β [9]. Nevertheless due<br />

to rapid production of neutralizing antibody against adenovirus, no<br />

improvement of gene transfer efficacy was achieved after the second dose<br />

[10]. The same research group conducted a clinical trial assessing the safety of<br />

adenoviral-mediated IFN-α2b in combination with chemotherapy. Recent<br />

data from this trial showed that the treatment is well tolerated and 25% of<br />

patients had partial response [11]. An intrapleural treament with re-directed T<br />

cells genetically engineered to express chimeric antigen receptor (CAR) that<br />

specifically recognizes tumor antigens is an attractive therapeutic option. A<br />

clinical phase I trial for intrapleural administration of fibroblast activation<br />

protein (FAP)-specific re-directed T cells is currently being conducted<br />

(NCT01722149). Photodynamic therapy (PDT) PDT is a light based cancer<br />

therapy. Most modern PDT applications involve three key components: a<br />

photosensitizer, a light source and tissue oxygen. The photosensitizing agent<br />

accumulates in tumor cells and is activated by light of a specific wavelength to<br />

produce reactive singlet oxygen that mediates cellular toxicity. The tumor<br />

cells are killed through both apoptosis and necrosis and by damaging tumor<br />

vasculature. It may also induce inflammatory reaction capable of stimulating<br />

a tumor directed host immune response. The advantages of this treatment are<br />

that its efficacy is not influenced by chemo- or radio-resistance of tumor cells,<br />

that it can be repeated at the same site without compromising its efficacy and<br />

that it does not compromise the ability to administer other treatment<br />

modalities in patients with recurrent or residual disease. PDT should be<br />

combined with macroscopic complete resection due to limited depth of<br />

penetration. Localized inflammation and fluid accumulation after treatment<br />

can modestly extend hospital stay. PDT appears promising and may improve<br />

local control and potentially prolong survival in properly selected patients<br />

who are able to undergo MCR, with clinical outcomes appearing best when<br />

PDT is combined with lung-sparing definitive surgery [12]. Friedberg reported<br />

a median survival of 31.7 months (41.2 months in patients with epithelioid<br />

histological subtype), but the progression free survival was only 9.6 months<br />

[13]. Intracavitary gene therapy (iGT) Gene therapy is based upon transfer of<br />

genetic material, including complementary DNA, full-lengths genes, small<br />

interfering RNA or oligonucleotids into cells for therapeutic purposes. For<br />

sufficient gene delivery, adenovirus is the most widely used in clinical trials<br />

among a variety of viral and non-viral vectors. In addition to delivering<br />

cytokine expressing vectors or re-directed T cells (see iIT part), several<br />

different cancer gene therapy approaches are currently used including the so<br />

called suicide gene therapy wherein a neoplasm is transduced with a cDNA<br />

encoding for an enzyme rendering tumor cells sensitive to a benign agent by<br />

converting the product to a toxic metabolite. The Herpes Simplex Virus 1-<br />

thymidine kinase (HSVtk) gene encodes for an enzyme that converts<br />

ganciclovir, an antiviral drug, to its cytotoxic metabolite. Intrapleural<br />

adenovirus HSVtk/ganciclovir administration was safe in MPM and two<br />

patients survived >6.5 years. Nevertheless, due to the fact that transgenes<br />

HSVtk were only detected at the surface of tumor tissues, the authors<br />

suggested that the treatment efficacy may be a result of antitumor immune<br />

response stimulation [14]. MPM tumor genome is characterized by frequent<br />

mutations in tumor suppressor genes such NF2, BAP1 or p53, thus the delivery<br />

of tumor suppressor gene expressing vectors into tumor cells can serve as an<br />

attractive treatment approach. The delivery of adenovirus expressing p53 has<br />

been tested in clinical trials for lung cancer but did not show better clinical<br />

benefit over chemotherapy [15]. This may be due to limited transfection<br />

efficiency of the vector and the stimulation of neutralizing antibody,<br />

therefore an improvement of transfection is still needed for the further<br />

development of gene therapy.References: 1. Sugarbaker, P.H., et al., Update on<br />

chemotherapeutic agents utilized for perioperative intraperitoneal<br />

chemotherapy. Oncologist, 2005. 10(2): p. 112-22. 2. Richards, W.G., et al.,<br />

Phase I to II Study of Pleurectomy/Decortication and Intraoperative<br />

Intracavitary Hyperthermic Cisplatin Lavage for Mesothelioma. J Clin Oncol,<br />

2006. 24(10): p. 1561-1567. 3. de Bree, E., et al., Cytoreductive surgery and<br />

intraoperative hyperthermic intrathoracic chemotherapy in patients with<br />

malignant pleural mesothelioma or pleural metastases of thymoma. Chest,<br />

2002. 121(2): p. 480-7. 4. Sugarbaker, D.J., et al., Hyperthermic intraoperative<br />

pleural cisplatin chemotherapy extends interval to recurrence and survival<br />

among low-risk patients with malignant pleural mesothelioma undergoing<br />

surgical macroscopic complete resection. J Thorac Cardiovasc Surg, 2013.<br />

145(4): p. 955-63. 5. Lardinois, D., et al., Intrapleural topical application of<br />

cisplatin with the surgical carrier Vivostat increases the local drug<br />

concentration in an immune-competent rat model with malignant<br />

pleuromesothelioma. J Thorac Cardiovasc Surg, 2006. 131(3): p. 697-703. 6.<br />

Tada, Y., et al., An intrapleural administration of zoledronic acid for inoperable<br />

malignant mesothelioma patients: a phase I clinical study protocol.<br />

Springerplus, 2016. 5: p. 195. 7. Astoul, P., et al., Intrapleural recombinant IL-2 in<br />

passive immunotherapy for malignant pleural effusion. Chest, 1993. 103(1): p.<br />

209-13. 8. Antoniou, K.M., E. Ferdoutsis, and D. Bouros, Interferons and their<br />

application in the diseases of the lung. Chest, 2003. 123(1): p. 209-16. 9.<br />

Sterman, D.H., et al., A phase I clinical trial of single-dose intrapleural IFN-beta<br />

gene transfer for malignant pleural mesothelioma and metastatic pleural<br />

effusions: high rate of antitumor immune responses. Clin Cancer Res, 2007.<br />

13(15 Pt 1): p. 4456-66. 10. Sterman, D.H., et al., A phase I trial of repeated<br />

intrapleural adenoviral-mediated interferon-beta gene transfer for<br />

mesothelioma and metastatic pleural effusions. Mol Ther, 2010. 18(4): p.<br />

852-60. 11. Sterman, D.H., et al., Pilot and Feasibility Trial Evaluating<br />

Immuno-Gene Therapy of Malignant Mesothelioma Using Intrapleural Delivery<br />

of Adenovirus-IFNalpha Combined with Chemotherapy. Clin Cancer Res, 2016.<br />

22(15): p. 3791-800. 12. Simone, C.B., 2nd and K.A. Cengel, Photodynamic<br />

therapy for lung cancer and malignant pleural mesothelioma. Semin Oncol,<br />

2014. 41(6): p. 820-30. 13. Friedberg, J.S., et al., Radical pleurectomy and<br />

intraoperative photodynamic therapy for malignant pleural mesothelioma.<br />

Ann Thorac Surg, 2012. 93(5): p. 1658-65; discussion 1665-7. 14. Sterman, D.H.,<br />

et al., Long-term Follow-up of Patients with Malignant Pleural Mesothelioma<br />

Receiving High-Dose Adenovirus Herpes Simplex Thymidine Kinase/Ganciclovir<br />

Suicide Gene Therapy. Clin Cancer Res, 2005. 11(20): p. 7444-7453. 15. Schuler,<br />

M., et al., Adenovirus-mediated wild-type p53 gene transfer in patients<br />

receiving chemotherapy for advanced non-small-cell lung cancer: results of a<br />

multicenter phase II study. J Clin Oncol, 2001. 19(6): p. 1750-8.<br />

Keywords: malignant pleural mesothelioma, intracavitary chemotherapy,<br />

Intracavitary Immunotherapy, photodynamic therapy<br />

SC06: NOVEL THERAPIES IN MALIGNANT PLEURAL MESOTHELIOMA AND THYMIC<br />

MALIGNANCIES<br />

MONDAY, DECEMBER 5, 2016 - 14:30-15:45<br />

SC06.04 IMMUNOTHERAPY OF MALIGNANT PLEURAL<br />

MESOTHELIOMA AND THYMIC MALIGNANCIES: THE END OF THE<br />

BEGINNING?<br />

Jan Van Meerbeeck<br />

<strong>Thoracic</strong> <strong>Oncology</strong>, Antwerp University Hospital, Edegem/Belgium<br />

The significant improvement in outcome observed with immune checkpoint<br />

S46 <strong>Journal</strong> of <strong>Thoracic</strong> <strong>Oncology</strong> • Volume 12 Issue S1 January 2017

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!