578349

research-article2015

TAR0010.1177/1753465815578349Therapeutic Advances in Respiratory DiseaseN Guibert, M Delaunay

Therapeutic Advances in Respiratory Disease

Review

Targeting the immune system to treat lung cancer: rationale and clinical experience Nicolas Guibert, Myriam Delaunay and Julien Mazières

Ther Adv Respir Dis 1­–16 DOI: 10.1177/ 1753465815578349 © The Author(s), 2015. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav

Abstract:  The use of immunotherapy that harnesses and enhances the innate powers of the immune system to fight cancer cells represents the most promising new cancer treatment approach since the development of the first chemotherapies and, more recently, targeted therapies. Unexpectedly, lung cancer has recently emerged as an exciting new target for immune-based therapies. Several approaches to immunotherapy for lung cancer have shown promise in early clinical trials and in late-phase development. The most advanced strategies can be split into two main categories: therapeutic vaccines and checkpoint inhibitors. At this time of great expectations, this review provides the reader with an update on the immunotherapies used to treat lung cancer with a focus on the rationale of targeting the immune system. It reports the results from recent major clinical trials, describes new toxicity profiles associated with such drugs, and particularly the role of the pulmonologists in their management. This review provides an overview of the main perspectives within this field. Keywords:  Immunotherapy, lung cancer, vaccine Introduction Activation of the immune system to treat cancer has long been investigated. After decades of disappointment, the tide has now finally changed due to the success of recent phase I clinical trials, especially for diseases with limited therapeutic options, such as melanoma and lung cancer. Durable responses have been recently reported for a broad range of human cancers using different agents that target the immune checkpoints [Gettinger and Herbst, 2014; Hamid and Carvajal, 2013; Hodi et  al. 2010; Topalian et al. 2012]. In addition to their promising activities, these treatments are usually well tolerated. We are currently in a transition period in treating lung cancer with immunotherapy: there are encouraging phase I–II trials, and numerous ongoing phase III clinical trials, but only one drug gets a recent approval in the USA. In this time of great expectations, we have aimed for this review to provide the reader with an update on the use of immunotherapy to treat lung cancer, with a focus on the rationale of targeting the immune system. We describe results obtained from major clinical trials, the new toxicity profiles

associated with such drugs, and particularly the role of pulmonologists in their management. We also provide a comprehensive overview of the main perspectives within this field. Rational for targeting the immune system in lung cancer It is somehow unexpected that, besides melanoma, the most promising results with immunotherapy to treat cancer have been observed in lung cancer. We argue that many features of lung cancer support these findings (Figure 1).

Correspondence to: Julien Mazières, MD, PhD Thoracic Oncology Unit, Respiratory Disease Department, Hôpital Larrey, CHU Toulouse, Chemin de Pouvourville, 31059 Toulouse Cedex, France mazieres.j@chu-toulouse. fr Nicolas Guibert, MD Myriam Delaunay, MD Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier, Toulouse, France

First, the lungs are the sanctuary of many immune disorders, along with the skin and kidneys (interestingly, all three organs being the best candidates for anticancer immunotherapy). Most autoimmune pathologies, such as disseminated lupus, Wegener’s disease, and other vascularitis are associated with lung complications. Pulmonary fibrosis is also a frequent disease linked to immune escape. Even more frequent diseases, such as asthma or chronic obstructive pulmonary disease, are associated, at least in part, with immune disorders, such as an imbalance in the T helper type 1 (Th1)/Th2 response.

http://tar.sagepub.com 1

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  (a)

Spontaneous resolution

(c)

Alveolar interface

(b)

Autoimmune diseases

(d)

PDL-1 expression

Figure 1.  Rationale for targeting the immune system in lung cancer. (a) Example of spontaneous regression of lung cancer [Gladwish et al. 2010]. (b) Example of usual interstitial pneumonia showing the frequency of interstitial immune-related disease of the lung. (c) The thin interface between the bronchoalveolar compartment and intrinsic immune regulators. (d) PDL1 expression in lung cancer. PDL1, programmed cell death ligand 1.

Second, the lungs are rich in various types of immunity cells, probably due to their chronic exposure to numerous external agents and the subsequent inflammation caused [Dasanu et  al. 2012]. Among these cells, regulatory T cells have been shown to play a major role in immune surveillance and immune adoptive responses. Third, the unique interface between the bronchoalveolar compartment and intrinsic immune regulators may play an important role in facilitating inflammatory alterations that promote lung cancer. Lung oncogenesis is marked by a complex array of immune defects, such as important T-cell alterations, natural killer cell dysfunction, B-lymphocyte defects, and defective dendritic cell, neutrophil, and monocyte functions [Sterlacci et al. 2012; Wesselius et al. 1987; Zikos et al. 2011]. Fourth, antigens are frequently expressed in lung tumor cells, some of which are shared with normal tissues, whereas others are unique to tumors. Tumor-specific antigens, such as angiotensinconverting enzyme, KRas, p53, and human telomerase reverse transcriptase, are frequently overexpressed or mutated in lung cancer [Hilbe et al. 2004] and are known to induce an immune response.

Lastly, some clinical reports of spontaneous resolution of lung cancer (as observed in patients with melanoma) suggest the importance of the immune response in tumor control [Gladwish et al. 2010]. Main mechanisms The main research findings supporting the use of immunotherapy in cancer are extensively reported in very elegant reviews [Chen and Mellman, 2013; Mellman et al. 2011]. We focus here on the steps of the cancer immunity cycle that can be targeted in lung cancer and the main mechanisms set up by the tumor to escape the immune response. Immunotherapy is considered active when it acts directly on the immune system and passive when its antitumor activity is based on an immunological mechanism. Active immunotherapy Active immunotherapy in oncology can be split into two main categories: those whose antitumor activity is antigen dependent and those that target immunological checkpoints. Treatments based on cytokines to enhance antitumor immune response are just mentioned here.

2 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. Antigen-dependent immunotherapy The purpose of the tumor-associated antigen (TAA) vaccines is to promote an antitumor immune response by enhancing the presentation of TAAs by dendritic cells to naïve T cells Major histocompatibility complex/T cell receptor (MHC–TCR interaction). This is the most widely studied immunotherapy. Yet the results are still disappointing (see below for details of the clinical studies). This is due to the different barriers related to the complexity of the immune response. The absence of dendritic cells that activate an adjuvant is one of the main reasons: the best adjuvant has still not been determined [granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin 2 (IL2) being the most widely used]. Determination of the best TAA candidate appears to be very tricky. The ideal antigen should be uniformly expressed on tumor cells but be missing from normal cells. It is not yet established whether the vaccine should be monovalent (with the risk of secondary resistance antigenic escape) or plurivalent. Another limiting factor is that the antigen may not be recognized or be recognized as a self antigen and thus induce a tolerogenic response [Motz and Coukos, 2013]. The tumor cell may also evade the immune system through the loss of expression of tumor antigens or MHC [Marincola et al. 2000; Whiteside, 2010]. Lastly, immune surveillance or artificial antigen-based immunotherapy can lead to selective survival of tumor cells that lack immunogenic epitopes, a process known as immunoediting [Schreiber et al. 2011]. Antigen-independent active immunotherapy: immune checkpoint blockade Immune checkpoint mechanisms normally prevent excessive and uncontrolled immune responses [Nirschl and Drake, 2013]. Briefly, TAAs must be directly presented by tumor cells or captured, processed, and presented by dendritic cells. These latter can, in turn, differentiate, migrate to the lymph nodes, and present TAA to naive T cells. The next step involves expansion of T cells in sufficient numbers to recognize and eliminate tumor cells. Antigeneducated T cells then leave the lymph node, travel to infiltrate the tumor, and eventually persist for enough time to kill the malignant cells. Immune checkpoints are crucial in this tumor immunity cycle and thus constitute potential targets for anticancer drugs. The priming of T-cell activation can be targeted by anticytotoxic T lymphocyte associated

protein 4 (CTLA4) antibodies, such as ipilimumab or tremelimumab, which block the interaction of the major negative regulator of T cells (CTLA4) with its ligands B7.1 and B7.2 (CD80 and CD86) [Qureshi et al. 2011]. CTLA4/CD80-86 interaction leads to anergy or redirects the T cells to a tolerogenic phenotype CD25-Foxp3 (T-regulatory cells), representing an ideal way for the tumor to escape the immune system [Darrasse-Jèze et al. 2009; Steinman et al. 2000, 2003].The identification of programmed cell death ligand 1 (PDL1) as a distal immune modulator, expressed in 20–50% of human cancers [Gettinger and Herbst, 2014], has also led to the development of a number of cancer immunotherapies that target the interactions between PD1 (programmed cell death 1) and PDL1, PDL1/B7.1, and PDL2/PD1 [Chen et al. 2012; Topalian et al. 2012]. Anti-CTLA4 monoclonal antibodies thus enable activation and expansion of tumor-infiltrating lymphocytes with a cytotoxic antitumor phenotype. This strategy’s main disadvantage is its obvious lack of selectivity and, therefore, the potential expansion of autoreactive lymphocytes that can induce autoimmune toxicity [Fong and Small, 2008]. The PDL1/2/PD1 pathway is a more distal secondary inhibitory immune checkpoint [Chen, 2004]. The transmembrane molecule PD1 is found on the surface of T lymphocytes, B lymphocytes, and monocytes [Keir et  al. 2008]. PDL1 is expressed either on tumor cells or on tumor-infiltrating immune cells. Their interaction causes the recruitment of the phosphatase Src-Homology Protein 2 (SHP2), subsequent inactivation of the PI3K/AKT signaling cascade, a change in the T-cell transcriptional profile, including blocking the secretion of cytotoxic mediators [Parry et al. 2005]. This pathway represents another immune escape mechanism for the tumor [Taube et al. 2012]. Passive immunotherapy: dendritic-cell-based immunotherapy Dendritic cells may acquire different phenotypes: proinflammatory when they secrete IL15, inducing a cytotoxic CTL response, and conversely more tolerogenic when they secrete interferon α (INFα) to induce a tolerogenic protumoral response mediated by INFγ and IL10 secretion [Banchereau and Palucka, 2005]. Proinflammatory dendritic cells can be generated ex vivo from patients’ monocytes, loaded with

http://tar.sagepub.com 3

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  TAAs, and then activated and injected into patients [Palucka and Banchereau, 2013]. Dendriticcell-based vaccines induce expansion of circulating CD4+ and CD8+ T cells, which are specific for tumor antigens. Passive immunotherapy: adoptive transfer of ex-vivo-activated T cells Chimeric antigen receptors are composed of a synthetic antibody against a tumor-surface antigen (with CD19 for B-cell malignancies being the most widely studied) fused to T-cell signaling domains [Kalos and June, 2013]. Genetically modified autologous transfected T cells are then reinfused into patients [Kochenderfer and Rosenberg, 2013]. This strategy still needs to be extended to cancers beyond hematologic malignancies. Vaccines: clinical results Antigen-specific active immunotherapies have been tested in several clinical trials with, up to now, disappointing results. Most antigen-specific immunotherapies have incorporated a single antigen and have narrow epitope specificities, which may contribute to their lack of efficacy in some trials [Slingluff, 2011]. We briefly report on the main trials below. Targeting MUC1 (Mucin 1) The phase III START trial [Butts et  al. 2014] randomized more than 1500 patients with unresectable stage III non-small cell lung cancer (NSCLC), previously treated with chemoradiotherapy, between treatment with tecemotide (a therapeutic vaccine targeting cancer cells expressing MUC1) or a placebo. Tecemotide failed to significantly prolong survival in the overall population. However, in a preplanned subgroup analysis, tecemotide improved survival in patients who had received concurrent chemoradiotherapy [30.8 months versus 20.6 months; hazard ratio (HR) 0.78, p = 0.016]. A further phase III trial is being initiated in this population (START2). The TG4010 vaccine also targets MUC1, but consists of a recombinant vaccinia virus encoding MUC1 and IL2. In a phase II study on advanced stage NSCLC, a slight albeit nonsignificant difference in progression-free survival (PFS) at 6 months was observed (43% with TG4010 combined with chemotherapy versus 35% with chemotherapy alone, p = 0.3) and no difference in terms of overall survival (OS) (10.7 months versus 10.3 months;

p = 0.59). A phase IIB–III study of TG4010 added chemotherapy to stage IV NSCLC (TIME trial): it has recently shown that TG4010 was efficacious and had an acceptable safety profile in patients with stage IV NSCLC, particularly in the nonsquamous population [Vansteenkiste et  al. 2013; Quoix et al. 2014]. Moreover, exploratory analysis of lymphocyte phenotypes at baseline performed in patients with evaluable samples suggested that the percentage of CD16+CD56+CD69+ cells, a phenotype of activated natural killer cells, was a potential predictor of outcome in patients who received TG4010 [Quoix et al. 2011]. Targeting melanoma-associated antigen 3 GSK1572932A is a vaccine that combines a melanoma-associated antigen 3 (MAGE-A3) peptide with the immune adjuvant AS15. A phase II study in patients with resected MAGE-A3+ NSCLC initially suggested a trend towards improved outcomes [Vansteenkiste et  al. 2013; Quoix et  al. 2014]. Unfortunately, these results were not confirmed in a recent large phase III study (MAGRIT trial), which showed no difference in terms of disease-free survival and no clear impact of the biomarkers [Vansteenkiste et al. 2014]. Targeting human telomerase reverse transcriptase GV1001, a peptide vaccine that corresponds to the active site of human telomerase reverse transcriptase and GM-CSF, induced specific immune responses in 80% of patients with unresectable stage III NSCLC [Brunsvig et al. 2011]. A gain in median OS was reported among immune responders (19.0 months versus 3.5 months for immune nonresponders, p < 0.001) in a phase I/II study in which GV1001 was combined with a second telomerase peptide (I540). A phase III study is ongoing. Other strategies The CIMAvax epidermal growth factor vaccine, developed in Cuba, is designed to induce an antibody-mediated immune response [Vinageras et al. 2008]. However, a phase II study failed to demonstrate a gain in median OS, but a subgroup of patients with a good antibody response appeared to derive some benefit from the drug (11.7 months versus 3.6 months, p = 0.002). The belagenpumatucel-L vaccine (LucanixTM, NovaTx Corporation, San Diego, California, USA)

4 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. consists of NSCLC cell lines transfected with a transforming growth factor β2 antisense gene. Despite a promising phase II study [Nemunaitis et al. 2006], a recent phase III trial on advancedstage NSCLC reported no significant difference in OS compared with a placebo in the overall study population, but with a potential benefit in subgroups with adenocarcinoma and who had been previously treated with radiotherapy [Giacone, 2013]. Tergenpumatucel-L (lung-cancer cell lines transfected with a murine galactosyl-transferase gene) was associated with median OS of 11.3 months in a phase II study that included 28 patients with metastatic or recurrent NSCLC. Induction of IFNγ secretion by antigen-specific T lymphocytes was associated with improved OS (21.9 months versus 7.2 months, p = 0.044) [Morris et al. 2013]. Altogether, the results are disappointing to date but subgroups appeared to derive more benefit than other groups and new trials are currently being conducting in selected patients.

versus 8.3 months with a placebo), but the difference in OS was not significant (p = 0.23). The PFS advantage was not observed in the concurrent ipilimumab arm [Lynch et al. 2012]. Two phase III studies [ClinicalTrials.gov identifier: NCT01285609 and NCT01450761] have recently evaluated ipilimumab in combination with paclitaxel–carboplatin or platinum–etoposide in patients with squamous NSCLC and small cell lung cancer. Results are awaited. Other trials are currently ongoing, such as a two phase I trials on ipilimumab plus targeted therapies for patients with stage IV NSCLC with epidermal growth factor receptor (EGFR) or ALK mutations [ClinicalTrials.gov identifier: NCT01998126]. Another trial is assessing ipilimumab plus imatinib [ClinicalTrials.gov identifier: NCT01738139], and a phase II neoadjuvant trial is assessing ipilimumab plus chemotherapy [ClinicalTrials.gov identifier: NCT01820754].

Targeting CTLA4 CTLA4 expression is not only confined to T lymphocytes but is also expressed in NSCLC tumors in 51–87% of cases. In one study, CTLA4 was associated with nonsquamous histology but was not prognostic for OS [Salvi et  al. 2012]. In another study, CTLA4 tumor expression was associated with older age and poor tumor differentiation [Sundar et al. 2014; Zheng et al. 2010].

A second antibody against CTLA4, tremelimumab, is a human immunoglobulin G2 (IgG2) monoclonal antibody. This antibody has been studied in a phase II open-label trial on advanced malignant mesothelioma in which the patients were chemotherapy resistant. In the tremelimumab arm, the median PFS was 6·2 months [95% confidence interval (CI) 1.3–11.1] and the median OS was 10.7 months (95% CI 0.0–21.9). A phase IIb, randomized, double-blind study that is comparing tremelimumab with a placebo for the second- or third-line treatment of subjects with unresectable pleural or peritoneal malignant mesothelioma is ongoing.

Ipilimumab Ipilimumab is an anticytotoxic T-cell lymphocyte 4 monoclonal antibody that was approved, in 2011 by the US Food and Drug Administration (FDA) to treat all patients with unresectable or metastatic melanoma. It was also approved, in 2013, by the European Medicines Agency for adult patients with previously treated advanced melanoma. Ipilimumab has been evaluated in a stage IIIb/IV NSCLC phase II study in combination with paclitaxel and carboplatin. Patients were randomized to receive either paclitaxel with carboplatin and a placebo, or concurrent ipilimumab or phased ipilimumab (frontline chemotherapy alone and subsequent combination of both regimens). Phased ipilimumab improved the immune-related PFS (5.7 months versus 4.6 months with a placebo) (HR 0.72, p = 0.05) and median OS (12.2 months

Targeting the PD1/PDL1 pathway PDL1 is a distal modulator of the immune response whose expression occurs in 40–50% of NSCLC cases [McLaughlin et  al. 2014; Yang et al. 2014]. Its prevalence is higher in men with squamous cell carcinoma at an advanced stage [McLaughlin et  al. 2014]. Although no association has been established with the main molecular biomarkers in stage I resected lung adenocarcinoma [Yang et  al. 2014], recent data suggest a correlation between tobacco and the KRAS mutation (OR 2.5) among all stages of combined populations [Ansen et  al. 2014; Calles et  al. 2014]. The main molecules currently under investigation are summarized in Table 1. The outcomes and toxicities observed with different molecules targeting PDL1/PD1 pathways are summarized in Table 2. Figure 3 shows the analysis of PDL1

http://tar.sagepub.com 5

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  Table 1.  Overview of the main checkpoint inhibitors developed in lung cancer Type

Name

Company

Trials (lung)

Anti-CTLA4  

Ipilimumab Tremelimumab

Phase III Phase II

Anti-PD1   Anti-PDL1

Nivolumab Pembrolizumab (MK3475) MEDI4736

BMS MedImmune, Astra-Zeneca BMS Merck

   

BMS936559 MPDL3280A

Phase II, III Phase III

MedImmune, Astra-Zeneca BMS Genentech Roche

Phase II, III Phase I Phase II, III

CTLA4, cytotoxic T lymphocyte associated protein 4.

Table 2.  Efficacy and toxicity observed with the main checkpoint inhibitors in lung cancer. Type

Name

n

RR

PFS (months)

OS (months)

AE all grade

Anti-PD1

Nivolumab

129

17%

2.3

9.9

53%

5%



Pembrolizumab MEDI4736 MPDL3280A

236

21%

2.5

8.2

48%

6%

58 53

16% 23%

NR 45% (at 6 months)

NR NR

34% 66%

0% 11%

Anti-PDL1  

AE grade 3–4

Pneumonia Grade 3–4: 2% Grade 3–4: 1% None None

AE, adverse event; NR, not reported; OS, overall survival; PD1, programmed cell death 1; PDL1, programmed cell death ligand 1; PFS, progression-free survival; RR, relative risk.

expression according to companies developing checkpoint inhibitors. Figure 4 shows two individual cases of tumor responses and toxicities after treatment with anti-PDL1 inhibitors. Anti-PD1 Nivolumab, an anti-PD1 IgG4 monoclonal antibody, is the first and most extensively evaluated anti-PD1 inhibitor of NSCLC. Results from a phase Ib study on nivolumab have reported objective response rates for pretreated NSCLC of 17% [Topalian et  al. 2012]. Median OS was 9.9 months, and was uninfluenced by histology (9.2 for squamous cell histology and 10.1 for a nonsquamous histology). It is important to note that efficacy appears sustainable (74 weeks median response duration, 1-year OS 42%, 2-year OS 24%) [Brahmer et al. 2014]. The best results were obtained at the dose of 3 mg/kg (1-year OS 50%, 2-year OS 45%) [Brahmer et al. 2014]. A recent update shows a 3-year survival rate of 17% for all patients and 27% for patients

receiving the 3 mg/kg dose [Gettinger and Herbst, 2014]. Given frontline as a monotherapy, at a dose of 3 mg/kg every 2 weeks, a response rate of 21% was observed, and this was higher in smokers and in patients harboring expression of PDL1 [Rizvi et al. 2014]. Median PFS was 15.6 weeks. As a first-line therapy, at a dose of 5 mg/kg, disease control rate was 51%, 71%, and 38%, when combined with gemcitabine–cisplatin, pemetrexed–cisplatin, and paclitaxel–carboplatin, respectively [Scott et al. 2014]. A recent phase II study (CA209-063) was conducted in pretreated patients with squamous-cell lung carcinoma [Rizvi et al. Lancet Oncol 2015]. Nivolumab, as a monotherapy, showed clinically meaningful efficacy in this population with an overall response rate (ORR) of 15%, durable responses (59% ongoing), and a 1-year OS of 41%. Clinical activity was observed in both PDL1– and PDL1+ patients with a higher although not significant benefit for the PDL1+ patients (ORR 24% versus 14%). Nivolumab was well tolerated with a manageable safety profile,

6 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. Immune cells expression

Tumor membrane expression PDL1 strong > 50%

Strong + > 5% PDL1 + (> 1%)

+

PDL1 weak+ > 1%

-

PDL1 -

PDL1 -

IHC3 > 10%

M2 > 50%

+

+

M2 > 25%

IHC1 > 1%

M1 > 1%

-

IHC2 > 5%

-

IHC0

+ -

M0 < 1%

Figure 2.  Legend of table 2.

New or worsening pulmonary symptoms (cough, dyspnea)

CT scan with contrast agents.

If clinically indicated discuss biology (hemogram, CRP), cardiac echography, spirometry

No change

Differenal diagnosis

(no addional features in comparison with baseline and last CT scan)

(pulmonary embolism, pulmonary infecon, tumor progression, others)

Differenal diagnosis

(infecon, tumor progression)

CT scan abnormalies

(infiltrates, patchy paerns, ground glass opacies, peribronchial consolidaon, intersal pneumonia)

Bronchoscopy

With Bronchiolo-Alveolar Lavage, cell count, T-cell subsets, virus, fungi, bacteria, Koch Bacillus, bronchial biopsies

Highly suspected iatrogenic pneumonia

  

Symptomac treatment Connuaon of immunotherapy Discuss repeat CT scan at 3 weeks.

 



Treatment according to eology. Discuss immunotherapy connuaon according to paent’s condion. Discuss repeat CT scan at 3 weeks.

Mild symptoms  Delay immunotherapy, start prednisone (1–2 mg/kg). Repeat CT scan.  Daily monitoring of symptoms Severe symptoms  Permanent disconnuaon of immunotherapy,  high dose of steroids (1 g/day 3 days then taper),  O2.  Consider broad spectrum anbiocs and other immunosuppressive medicaons.

Figure 3.  Algorithm of management pulmonary symptoms in patient treated with immunotherapy.

but 12% of patients discontinued treatment due to drug toxicity (4% pneumonitis), and two treatment-related deaths (one hypoxic pneumonia and one ischemic stroke) occurred in patients with multiple comorbidities and concurrent progressive disease. Two phase III trials that

compared OS with nivolumab versus docetaxel in advanced, previously treated nonsquamous (CA209-057) [ClinicalTrials.gov identifier: NCT01673867] and squamous NSCLC have been recently completed (CA209-017) [ClinicalTrials.gov identifier: NCT01642004].

http://tar.sagepub.com 7

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease 

Figure 4.  Example of tumor response after treatment with programmed cell death ligand 1 inhibitor (a, b) and example of programmed cell death 1 inhibitor induced interstitial pneumonia (c) (all personal cases).

Pembrolizumab (MK-3475) is another IgG4 antiPD1 antibody. Pembrolizumab was approved by the FDA in September 2014 for the treatment of advanced melanoma and was granted a ‘breakthrough-therapy’ designation in October 2014 for the treatment of patients with pretreated NSCLC who were negative for EGFR and ALK. This study follows the encouraging intermediary results of the KEYNOTE-001 study (n = 307), which showed a response rate of 21% (RECIST) and 23% (immune-related response criteria) [Wolchok et al. 2009]. A median PFS of 27 months among treatment-naïve patients and 10 months among pretreated patients was observed. Again, the responses observed were usually durable [Garon et al. 2014]. Using a stringent test, with a positivity cutoff at 50% of cells expressing PDL1, a clinically relevant difference was observed in terms of response between the strong positive group (37%) and the negative group (10%). First-line and combination trials are ongoing, as follows. There is a phase III trial on pembrolizumab given to patients with PDL1+ NSCLC

[ClinicalTrials.gov identifier: NCT01905657], a phase II study on pembrolizumab given to patients with NSCLC and brain metastases [ClinicalTrials.gov identifier: NCT02085070], a phase I/II trial on pembrolizumab in combination with chemotherapy or a targeted therapy for patients with NSCLC [ClinicalTrials.gov identifier: NCT02039674], a phase I/II trial on pembrolizumab in combination with an IDO (indoleamine 2,3-dioxygenase) inhibitor given to patients with advanced NSCLC [ClinicalTrials. gov identifier: NCT02178722], and a phase I trial on pembrolizumab given to patients with biomarker-positive solid tumors, including lung cancer [ClinicalTrials.gov identifier: NCT02054806]. Anti-PDL1 MPDL3280A, a human anti-PDL1 monoclonal antibody, led to antitumor responses, with an ORR of 23% (12/53) in patients with previously treated NSCLC in a phase I study, with a rapid and durable response [Figure 4(a, b)] [Brahmer 2014]. Moreover, the treatment was well tolerated.

8 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. Patients with PDL1+ tumors showed an ORR of 39%, and a progressive disease rate of 12%. Patients with PDL1– tumors had an ORR of 13% with a progressive disease rate of 59%. These results suggested a correlation between a response to the agent and PDL1 status [Herbst et al. 2013; Spiegel et al. 2013]. Other one-arm or randomized phase II and III studies are ongoing. Following a phase II randomized trial (POPLAR) [ClinicalTrials.gov identifier: NCT01903993], a phase III trial on MPDL3280A (OAK) [ClinicalTrials.gov identifier: NCT02008227] has just been completed. This open-label, multicenter study is designed to evaluate the efficacy and safety of MPDL3280A versus docetaxel in patients with locally advanced or metastatic NSCLC who have failed a platinumbased chemotherapy. Two phase II trial are dedicated to patients with PDL1+, locally advanced or metastatic NSCLC (FIR and BIRCH) [ClinicalTrials.gov identifier: NCT01846416, NCT02031458]. A phase I trial is evaluating the combination of MPDL3280A combined with bevacizumab or bevacizumab plus chemotherapy for patients with advanced NSCLC [ClinicalTrials. gov identifier: NCT01633970]. MEDI4736 is another anti-PDL1 monoclonal antibody that achieved a 13% objective response rate and a 30% disease control rate in NSCLC, in a phase I study [Segal et  al. 2014]. Currently within recruitment, the ATLANTIC [ClinicalTrials.gov identifier: NCT02087423] is a single-arm, phase II global study on the efficacy and safety of MEDI4736 in pretreated patients with locally advanced or metastatic NSCLC. The PACIFIC trial [ClinicalTrials.gov identifier: NCT02125461] is a phase III study on MEDI4736 versus a placebo given to patients with locally advanced, unresectable NSCLC, with no evidence of progression after completion of treatment of chemoradiotherapy. A phase II/III trial on MEDI4736, as a second-line therapy, is also ongoing for patients with recurrent stage IIIB/IV NSCLC [ClinicalTrials.gov identifier: NCT02154490]. In addition, there are several early phase I studies on NSCLC using MEDI4736 in combination with gefitinib [ClinicalTrials.gov identifier: NCT02088112], tremelimumab [ClinicalTrials.gov identifier: NCT02000947], and MEDI0680 [ClinicalTrials.gov identifier: NCT02118337], an anti-PD1 monoclonal antibody (in advanced malignancies).

The different drugs targeting the CTLA4 and PD1/PDL1 pathways and the ongoing phase II/ III clinical trials are summarized in Table 1. Toxicity Targeting immune checkpoints induced the emergence of a new form of toxicity termed immunerelated adverse effects (irAEs). Due to its more distal localization in the cancer immunity cycle, inhibition of the PD1 pathway enhances the effector function of previously activated T cells. Conversely, CTLA4 blockade both promotes the activation of preexisting T cells and redirects naïve T cells into the cytotoxic phenotype. This difference theoretically suggests a lower risk of autoimmune disorders by targeting the PD1/ PDL1 pathway. Anti-PDL1 antibodies may also have better selectivity for tumor cells than antiPD1, with fewer irAEs [Zou and Chen, 2008]. The most commonly observed irAEs with all checkpoint inhibitors are the following: (1) Lung: pneumonitis is defined as a disorder characterized by focal or diffuse inflammation that affects the lung parenchyma. (2) Skin: rash or pruritus (grade 1) is frequent. The appearance of a rash may be explained by an ipilimumab-stimulated immune response to melanocytes [Weber et al. 2009]. This is supported by the appearance of vitiligo in 11% of patients in a phase II study on ipilimumab [Hodi et al. 2003]. Toxic epidermal necrolysis, as well as Stevens–Johnson syndrome (grade 3–4), are rarely reported. (3) Gastrointestinal: diarrhea and colitis are frequent, whereas hepatotoxicity and pancreatitis are uncommon. (4) Endocrine glands: hypophysitis and hypothyroidism have been reported. Anti-CTLA4 Blockade of CTLA4 signaling with ipilimumab or tremelimumab increases T-cell activation and restores T-cell proliferation. Part of the toxicity (irAEs) is probably induced by cytokines released by activated T cells. In a pooled analysis of 325 patients treated with 10 mg/kg ipilimumab, once every 3 weeks for four times: irAEs of any grade were observed in 72.3%. Grade 3 or 4 irAEs were observed in 25.2% of patients, mainly in the gastrointestinal tract (12%), liver (7%), skin (3%), and endocrine system (3%) [Weber et al. 2012].

http://tar.sagepub.com 9

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  Anti-PDL1 IrAEs, including mild fatigue, rash, diarrhea, and colitis, have been described with anti-PD1 agents, although adverse events seem to be less common than those seen with anti-CTLA4 antibodies [Gangadhar and Vonderheide, 2014]. Nivolumab was generally well tolerated with skin toxicities (20%), gastrointestinal (15%), and pulmonary (9%) adverse events being most commonly observed. A lower frequency of gastrointestinal toxicities was seen: 2% (grade 3/4) compared with 20% with ipilimumab. Pneumonitis was reported in 6% (8/129) [Brahmer et al. 2014]. A rare but potentially fatal inflammatory pneumonitis was observed in a few cases [Figure 4(c)], which led to three treatment-related deaths in the original phase I study on nivolumab [Topalian et  al. 2012] and one related death in the CHECKMATE 063 trial [Rizvi et al. Lancet Oncol 2015]. The other drugs, pembrolizumab, MEDI-4736, and MPDL3280A, have very similar safety profiles with mild pulmonary toxicities. The main toxicities observed with checkpoint inhibitors are summarized in Table 2.

If there is any doubt, a bronchoscopy should be considered with an oriented bronchoalveolar lavage that includes a cell count, analysis of T-cell subsets, microbial analysis (including viruses, fungi, bacteria and BK) and, according to the patient’s status, a transbronchial biopsy, which can help narrow down the diagnosis [Barjaktarevic et al. 2013; Rizvi et al. Lancet Oncol 2015]. Some recommendations have been proposed [Ramaligan et al. 2014] based on the following clinical signs.

Management of pulmonary toxicity Pulmonary toxicity needs to be recognized and treated early as it can be life threatening. Pneumonitis management has been described in clinical experience and observational reports but without consensus, standardization, or validation in prospective clinical trials [Inoue et  al. 2001; Lynch et  al. 2012]. It is unknown whether immune-related pneumonitis relates to other forms of described pneumonitis or how it should best be managed [Chow, 2013]. Other forms of severe or even fatal drug-related pneumonitis have been described in patients with NSCLC treated with erlotinib (1.6–4.5%) [Liu et  al. 2007], gefıtinib (3.5%) [Konishi et  al. 2005], docetaxel (4.6%) [Grande et  al. 2007], and gemcitabine (1–2%) [Roychowdhury et  al. 2002], with two case reports of pemetrexed-related pneumonitis [Hochstrasser et al. 2012].

Grade 3 (severe symptoms, limited self care or activities of daily living, or oxygen) and above, immunotherapy treatment should be discontinued permanently and high doses of intravenous steroids (for example: methylprednisolone 1 g/ day) administered in conjunction with oxygen and ventilatory support, if needed. Bronchoscopy or lung biopsies are indicated if clinically feasible. In cases of failure after 48 h or serious evolution, it should be considered to use broad-spectrum prophylactic antibiotics and additional immunosuppressive medication. In the protocol of some clinical trials addition of infliximab, mycophenolate mofetil, or cyclophosphamide is proposed even if data are lacking to validate the use of such drugs. Herein, we propose an algorithm to help manage the onset of pulmonary symptoms in patients treated with immunotherapy (Figure 3).

Patients with signs and symptoms of pneumonitis, such as dyspnea, cough, hypoxia, and interstitial syndrome (as observed on a chest X-ray) should undergo a computed tomography scan. This exam can confirm pneumonitis (extensive, patchy, bilateral, peribronchial consolidation) or orient towards a differential diagnosis (infection, heart failure, tumor progression, pulmonary embolism).

For grade 1 (asymptomatic, clinical, or diagnostic observations only, intervention not indicated): it is possible to discuss utilization of steroids and empiric antibiotics (if there is a suspicion of concurrent infection) and to repeat thoracic imaging 3 weeks later. For grade 2 (symptomatic and medical intervention is indicated, limited ability for daily living): delay immunotherapy dosing and start moderate doses (1–2 mg/kg) of prednisone, which is then slowly tapered over 4 weeks or more; a bronchoalveolar lavage with a bronchoscopy is also recommended.

Perspectives Combination of drugs Even though some immunotherapies may have activity as single agents, more impressive activity can be seen when they are combined with other agents. Combinations of immune therapies and chemotherapy agents are currently being studied in the setting of advanced disease. Accumulating

10 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. evidence suggests the rational for different drug associations: Chemotherapies and immunotherapies Several conventional chemotherapies that induce immunogenic tumor-cell death can enhance a strong adaptive immune response. Indeed, tumor cells are converted into an anticancer vaccine (release of endogenous tumor antigens), and their effects may be enhanced by immune checkpoint inhibitors [Vacchelli et al. 2014]. Numerous clinical trials are ongoing, and are testing such combinations in small cell lung cancer and NSCLC. Anti-PD1/PDL1, anti-CTLA4 antibodies, and antitumor vaccines Prolonged antigenic exposure leads to expression of PD1/PDL1 and T-cell anergy [Blackburn et al. 2009]. This constitutes a robust rational for a synergic combination of antigen-specific active immunotherapies with immune checkpoint inhibitors because CTLA4 and PD1 inhibit T-cell activation and clonal T expansion after vaccination. Anti-PD1/PDL1 and anti-CTLA4 antibodies Because of two distinct levels of intervention, that is, T-cell activation and expansion in the periphery of CTLA4 and CTL effector functions in the tumor microenvironment of PD1, it seems logical to combine these two drugs to enhance antitumor immune activity [Wolchock et  al. 2013]. The combination of ipilimumab plus nivolumab has shown response rates of 11–33% depending on histology, but induces frequent and sometimes severe irAEs (85% toxicity, 48% grade 3–4, three deaths among 46 patients) [Antonia et al. 2014]. Immunotherapy and targeted therapy Activating a patient’s immune system during a time of tumor reduction and remission may be the best way to ensure that responses are converted to a long-term and durable benefit. Unlike conventional chemotherapies, targeted therapies for EGFR-mutated or ALK-rearranged adenocarcinoma may achieve rapid and significant tumor shrinkage without the need for immunosuppression induced by chemotherapies. Moreover, oncogenic EGFR signaling remodels the tumor microenvironment to trigger an immune escape and mechanistically links the treatment response to PD1 inhibition [Akbay et al. 2013]. A first-line

therapy that combines nivolumab and erlotinib shows excellent response rates, even in EGFR– tyrosine kinase inhibitor (TKI) pretreated patients, but with a grade 3–4 adverse event incidence of 24% [Naiyer et al. 2014]. Other trials combining, for example, gefitinib and tremelimumab in patients with EGFR mutation who progress under EGFR–TKI are ongoing. Biomarkers As with other targeted therapies, only a minority of patients currently benefit from active immunotherapy even though, for some patients, the benefits can be substantial. Patient selection is therefore a crucial issue in the development of such drugs. Initial assessments of active immunotherapies have generally been in patients with advanced-stage disease. However, rapid tumor growth and extensive tumor-related immunosuppression suggest that these patients may be least likely to benefit from this treatment. Ipilimumab Among patients treated with ipilimumab, an early increase in lymphocyte and eosinophil counts is associated with improved survival but no robust pretreatment biomarker has yet been identified [Delyon et al. 2013]. Anti-PD1 and anti-PDL1 antibodies Strong arguments now exist to consider PDL1 expression in immunohistochemistry (IHC) analysis as a potential predictive biomarker for the response to drugs that target the PDL1/PD1 pathway. In the MPDL3280A phase I study, ORR was 46% (6/13) in patients with a PDL1 IHC score of 2 or 3, and 83% (5/6) in those with a PDL1 IHC score of 3 [Horn et  al. 2013]. MEDI4736 response rate was also strongly correlated with PDL1 expression (39% versus 5% ORR) [Segal et  al. 2014]. Response rates in the KEYNOTE-001 study (pembrolizumab) were 37%, 17%, and 10% for strong positive, weak positive, or negative PDL1 expression; respectively [Garon et  al. 2014]. Our feeling is that PDL1 expression is not yet a perfect marker, as the optimal test has not been clearly defined and some patients who are PDL1– derive a benefit from these drugs even if the test is negative. Nevertheless, it appears to us to be an interesting basis for selecting patients, especially in the firstline setting, as chemotherapy (which can be

http://tar.sagepub.com 11

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  Table 3.  Biomarker test for PDL1 expression according to the companies. Name

Test

Analysis

Analyzed cells

Positivity

Expression

Nivolumab

Dako IHC (28-8 rabbit Ab)

FFPE archive

Tumor cell membrane expression

Cutoff at 1 and 5% > 100 tumor cells

Pembrolizumab

Dako IHC (22C3 mouse Ab)

FFPE archive

Tumor cell membrane expression

Cutoff at 50% (strong) and 1–49 (weak)

MEDI4736

Ventana IHC (SP263)

FFPE archive

Tumor cell membrane expression

MPDL3280A

Ventana IHC (SP142)

FFPE archive

Immune cell expression

M0 < 1% M1 > 1% M2 > 25% M3 > 50% IHC3 (>10%), IHC2 (>5%), IHC1 (> 1%)

Cutoff 1% : 56% Cutoff 5% : 49% Cutoff 50%: 25% Cutoff 1%: 70% Cutoff 25% M0, M1: 75% M2, M3: 25% IHC3: 11% IHC1: 75%

IHC, immunohistochemistry; FFPE, formalin-fixed, paraffin-embedded.

associated with bevacizumab and maintenance therapy) is associated with a response rate and a PFS that can reach 35% and 7.4 months [Barlesi et  al. 2013; Sandler et  al. 2006], respectively. Moreover, recent studies underlined the potential predictive factor of immune markers not only in the tumor cells but also in the surrounding immune cells. Tumeh and colleagues showed that CD8, PD1, and PDL1 expression obtained from pretherapeutic biopsies in the tumor and at the invasive margin significantly correlated with response to pembrolizumab in melanoma [Tumeh et al. 2014]. Similarly, in other studies across multiple cancer types, responses to MPDL3280A were observed in patients with tumors expressing high levels of PDL1, especially when PDL1 was expressed by tumor-infiltrating immune cells. Furthermore, responses were associated with Th1 gene expression, CTLA4 expression, and the absence of fractalkine (CX3CL1) in baseline tumor specimens [Herbst et  al. 2014]. We can thus anticipate that biomarkers might be dispensable in heavily pretreated patients who have no other therapeutic options, but will become mandatory in pretreated patients. The main characteristics of the PDL1 test used by different companies and the cutoff values are reported in Table 3 and Figure 3. Immunotherapy in the early stage of disease Initial assessments of active immunotherapies have been conducted in patients with advanced-stage disease. However, rapid tumor growth and extensive

tumor-related immunosuppression suggest that these patients might be least likely to benefit from this treatment. Another perspective in the field is thus to extend the indication of immunotherapy in the disease’s earlier stages. The 5-year survival for patients treated with surgery alone or with adjuvant chemotherapy remains low, underlining the need for new strategies. Moreover, not all patients with early-stage disease are eligible or willing to undergo chemotherapy following complete surgical resection. In the context of early-stage NSCLC, immunotherapeutic interventions that can both induce cell-mediated immunity against proliferating cancer cells following complete resection, and establish immunologic memory that may guard against future recurrences through active immune surveillance, are particularly attractive therapeutic options. Trials are about to begin in this setting (BR31-IFCT1401) [ClinicalTrials.gov identifier: NCT02273375]. Neoadjuvant trials are also interesting strategies that can initiate an immune response before surgery, even if the specific toxicity profile has to be addressed first in small phase I/II studies. Conclusion Enhancing the immune system in subjects with lung cancer represents an appealing therapeutic venue. As some immunotherapies have demonstrated significant activity in lung cancer, they may soon become a powerful addition to the oncologist’s toolbox. More basic research and clinical trials are needed to define which tumors

12 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. (histology, size, stage), which patients (age, pretreatments, smoking habits), and which combinations (alone, with chemotherapies, or targeted therapies) are the best candidates.

in non-small cell lung cancer (NSCLC): updated analysis of patients enrolled within 12 weeks of completion of chemotherapy. J Clin Oncol 32 (Suppl.): abstract 8056.

Conflict of interest statement The authors declare no conflicts of interest in preparing this article.

Blackburn, S., Shin, H., Haining, W., Zou, T., Workman, C., Polley, A. et al. (2009) Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10: 29–37.

Funding This research received no specific grant from any funding agency in the public, commercial, or notfor-profit sectors.

Brahmer, J. et al. (2013) Mini-oral presentation at WCLC 2013. J Thorac Oncol 8(Suppl. 2): abstract MO18.03.

References Akbay, E., Koyama, S., Carretero, J., Altabef, A., Tchaicha, J., Christensen, C. et al. (2013) Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov 3: 1355–1363. Ansen, S., Schultheis, A.M., Hellmich, M., Zander, T., Brockmann, M., Stoelben, E. et al. (2014) PD-L1 expression and genotype in non-small cell lung cancer. J Clin Oncol 32(Suppl.): abstract 7517. Antonia, S., Gettinger, S., Chow, L., Juergens, R., Borghaei, H., Shen, Y. et al. (2014) Nivolumab (anti-PD-1; BMS-936558, ONO-4538) and ipilimumab in firstline NSCLC: Interim phase I results. J Clin Oncol 32(Suppl.): abstract 8023. Antonia, S.J., Brahmer, J., Gettinger, S., Chow, L., Juergens, R., Shepherd, F.A. et al. (2014) Nivolumab (anti-PD-1; BMS- 936558, ONO4538) in combination with platinum based doublet chemotherapy (PT-DC) in advanced non-small cell lung cancer (NSCLC). J Clin Oncol 32(Suppl.): abstract 8113. Banchereau, J. and Palucka, A. (2005) Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol 5: 296–306. Barjaktarevic, I., Qadir, N., Suri, A., Santamauro, J. and Stover, D. (2013) Organizing pneumonia as a side effect of ipilimumab treatment of melanoma. Chest 143: 858–861. Barlesi, F., Scherpereel, A., Rittmeyer, A., Pazzola, A., Ferrer Tur, N., Kim, J. et al. (2013) Randomized phase III trial of maintenance bevacizumab with or without pemetrexed after first-line induction with bevacizumab, cisplatin, and pemetrexed in advanced nonsquamous non-small-cell lung cancer: AVAPERL (MO22089). J Clin Oncol 31: 3004–3011. Bazhenova, L., Giaccone, G., Nemunaitis, J., Juhász, E., Ramlau, R., Van den Heuvel, M. et al. (2014) An international, multicenter, randomized, double-blind phase III study of maintenance belagenpumatucel-l

Brahmer, J., Horn, L., Gandhi, L., Spigel, D., Antonia, S.J., Rizvi, N. et al. (2014) Nivolumab (anti-PD-1, BMS-936558, ONO-4538) in patients with advanced non-small-cell lung cancer (NSCLC): survival and clinical activity by subgroup analysis. J Clin Oncol 32(Suppl.): abstract 8112. Brunsvig, P., Kyte, J., Kersten, C., Sundstrøm, S., Møller, M., Nyakas, M. et al. (2011) Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial. Clin Cancer Res 17: 6847–6857. Butts, C., Socinski, M., Mitchell, P., Thatcher, N., Havel, L., Krzakowski, M. et al. (2014) Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol 15: 59–68. Calles, A. Liao, X., Sholl, L., Butaney, M., Rodig, S., Freeman, G.J. et al. (2014) Differential expression of LKB1, PD-L1, and PD-L2 in KRAS-mutant nonsmall cell lung cancer in never-smokers. J Clin Oncol 32(Suppl.): abstract 8032. Chen, D. and Mellman, I. (2013) Oncology meets immunology: the cancer-immunity cycle. Immunity 39: 1–10. Chen, D., Irving, B. and Hodi, F. (2012) Molecular pathways: next-generation immunotherapy–inhibiting programmed death-ligand 1 and programmed death1. Clin Cancer Res 18: 6580–6587. Chen, L. (2004) Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol 4: 336–347. Chow, L. (2013) Exploring novel immune-related toxicities and endpoints with immune-checkpoint inhibitors in non-small cell lung cancer. Am Soc Clin Oncol Educ Book. doi: 10.1200/EdBook_AM.2013.33. e280. Darrasse-Jèze, G., Deroubaix, S., Mouquet, H., Victora, G., Eisenreich, T., Yao, K. et al. (2009) Feedback control of regulatory T cell homeostasis by dendritic cells in vivo. J Exp Med 31 206: 1853–1862.

http://tar.sagepub.com 13

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  Dasanu, C., Sethi, N. and Ahmed, N. (2012) Immune alterations and emerging immunotherapeutic approaches in lung cancer. Expert Opin Biol Ther 12: 923–937.

Hochstrasser, A., Benz, G., Joerger, M., Templeton, A., Brutsche, M., Früh, M. (2012) Interstitial pneumonitis after treatment with pemetrexed: a rare event? Chemotherapy 58: 84–88.

Delyon, J., Mateus, C., Lefeuvre, D., Lanoy, E., Zitvogel, L., Chaput, N. et al. (2013) Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: an early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann Oncol 24: 1697–1703.

Hodi, F., Mihm, M., Soiffer, R., Haluska, F., Butler, M., Seiden, M. et al. (2003) Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA 100: 4712–4717.

Fong, L. and Small, E. (2008) Anti-cytotoxic T-lymphocyte antigen-4 antibody: the first in an emerging class of immunomodulatory antibodies for cancer treatment. J Clin Oncol 26: 5275–5283.

Hodi, F., O’Day, S., McDermott, D., Weber, R., Sosman, J., Haanen, J. et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363: 711–723.

Gangadhar, T. and Vonderheide, R. (2014) Mitigating the toxic effects of anticancer immunotherapy. Nat Rev Clin Oncol 11: 91–99.

Horn, L. et al. (2013) Mini-oral presentation at WCLC 2013. J Thorac Oncol 8(Suppl. 2): abstract MO18.01.

Garon, E.B., Gandhi, L., Rizvi, N., Hui, R., Balmanoukian, A.S., Patnaik, A. et al. (2014) Antitumor activity of pembrolizumab (Pembro; MK-3475) and correlation with programmed death ligand 1 (PD-L1) expression in a pooled analysis of patients (pts) with advanced non-small cell lung carcinoma. Ann Oncol 25(5): 1–41. doi: 10.1093/ annonc/mdu438.

Inoue, A., Kunitoh, H., Sekine, I., Sumi, M., Tokuuye, K. and Saijo, N. (2001) Radiation pneumonitis in lung cancer patients: a retrospective study of risk factors and the long-term prognosis. Int J Radiat Oncol Biol Phys 49: 649–655. Kalos, M. and June, C. (2013) Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity 39: 49–60.

Gettinger, S. and Herbst, R. (2014) B7-H1/PD-1 blockade therapy in non-small cell lung cancer: current status and future direction. Cancer J 20: 281–289.

Keir, M., Butte, M., Freeman, G. and Sharpe, A. (2008) PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26: 677–704.

Gladwish, A., Clarke, K. and Bezjak, A. (2010) Spontaneous regression in advanced non-small cell lung cancer. BMJ Case Rep. doi: 10.1136/ bcr.07.2010.3147.

Kochenderfer, J. and Rosenberg, S. (2013) Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat Rev Clin Oncol 10: 267–276.

Grande, C., Villanueva, M., Huidobro, G. and Casal, J. (2007) Docetaxel-induced interstitial pneumonitis following non-small-cell lung cancer treatment. Clin Transl Oncol 9: 578–581.

Konishi, J., Yamazaki, K., Kinoshita, I., Isobe, H., Ogura, S., Sekine, S. et al. (2005) Analysis of the response and toxicity to gefitinib of non-small cell lung cancer. Anticancer Res 25: 435–441.

Hamid, O. and Carvajal, R. (2013) Anti-programmed death-1 and anti-programmed death-ligand 1 antibodies in cancer therapy. Expert Opin Biol Ther 13: 847–861.

Liu, V., White, D., Zakowski, M., Travis, W., Kris, M., Ginsberg, M. et al. (2007) Pulmonary toxicity associated with erlotinib. Chest 132: 1042–1044.

Herbst, R., Gordon, M., Fine, G.D., Sosman, J.A., Soria, J.C., Hamid, O. et al. (2013) A study of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic tumors. J Clin Oncol 31(Suppl.): abstract 3000.

Lynch, T., Bondarenko, I., Luft, A., Serwatowski, P., Barlesi, F., Chacko, R. et al. (2012) Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol 30: 2046–2054.

Herbst, R., Soria, J., Kowanetz, M., Fine, G., Hamid, O., Gordon, M. et al. (2014) Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563–567.

Marincola, F., Jaffee, E., Hicklin, D. and Ferrone, S. (2000) Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 74: 181–273.

Hilbe, W., Dirnhofer, S., Greil, R. and Wöll, E. (2004) Biomarkers in non-small cell lung cancer prevention. Eur J Cancer Prev 13: 425–436.

McLaughlin, J., Schalper, K., Carvajal-Hausdorf, D., Velcheti, V., Haack, H., Silver, M. et al. (2014) Domain-specific PD-L1 protein measurement in

14 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

N Guibert, M Delaunay et al. non-small cell lung cancer (NSCLC). J Clin Oncol 32(Suppl.): abstract 8064.

for the cell-extrinsic function of CTLA-4. Science 332: 600–603.

Mellman, I., Coukos, G. and Dranoff, G. (2011) Cancer immunotherapy comes of age. Nature 480: 480–489.

Rizvi, N., Chow, L., Borghaei, H., Shen, Y., Harbison, C., Alaparthy, S. et al. (2014) Safety and response with nivolumab (anti-PD-1; BMS-936558, ONO-4538) plus erlotinib in patients with epidermal growth factor receptor mutant (EGFR MT) advanced NSCLC. J Clin Oncol 32(Suppl.): abstract 8022.

Morris, J., Rossi, G., Harold, N., Tennant, L., Ramsey, W., Vahanian, N. et al. (2013) Potential chemo-sensitization effect of tergenpumatucel-L immunotherapy in treated patients with advanced non-small cell lung cancer. J Clin Oncol 31(Suppl.): abstract 8094. Motz, G. and Coukos, G. (2013) Deciphering and reversing tumor immune suppression. Immunity 39: 61–73. Naiyer, A., Rizvi, N.A., Chow, L., Borghaei, H., Shen, Y., Harbison, C. et al. (2014) Safety and response with nivolumab (anti-PD-1; BMS-936558, ONO-4538) plus erlotinib in patients (pts) with epidermal growth factor receptor mutant (EGFR MT) advanced NSCLC. J Clin Oncol 32(Suppl.): abstract 8022. Nemunaitis, J., Dillman, R., Schwarzenberger, P., Senzer, N., Cunningham, C., Cutler, J. et al. (2006) Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J Clin Oncol 24: 4721–4730. Nirschl, C. and Drake, C. (2013) Molecular pathways: coexpression of immune checkpoint molecules: signaling pathways and implications for cancer immunotherapy. Clin Cancer Res 19: 4917–4924. Palucka, K. and Banchereau, J. (2013) Dendriticcell-based therapeutic cancer vaccines. Immunity 39: 38–48. Parry, R., Chemnitz, J., Frauwirth, K., Lanfranco, A., Braunstein, I., Kobayashi, S. et al. (2005) CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 25: 9543–9553. Quoix, E., Losonczy, G., Forget, F., Chouaid, C., Papai, Z., Gervais, R. et al. (2014) 1055PD TIME, a phase 2b/3 study evaluating TG4010 in combination with first line therapy in advanced non small cell lung cancer (NSCLC). Phase 2b results. ESMO: abstract 5152. Available at: https://www. webges.com/cslide/library/esmo/browse/search/8eQ” \l”9f9A02wQ”https://www.webges.com/cslide/library/ esmo/browse/search/8eQ#9f9A02wQ Quoix, E., Ramlau, R., Westeel, V., Papai, Z., Madroszyk, A., Riviere, A. et al. (2011) Therapeutic vaccination with TG4010 and first-line chemotherapy in advanced non-small-cell lung cancer: a controlled phase 2B trial. Lancet Oncol 12: 1125–1133. Qureshi, O., Zheng, Y., Nakamura, K., Attridge, K., Manzotti, C., Schmidt, E. et al. (2011) Transendocytosis of CD80 and CD86: a molecular basis

Rizvi, N.A., Mazières, J., Planchard, D., Stinchcombe, T.E., Dy, G.K., Antonia, S.J. et al. (2015) Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol 16(3): 257–265. Roychowdhury, D., Cassidy, C., Peterson, P. and Arning, M. (2002) A report on serious pulmonary toxicity associated with gemcitabine-based therapy. Invest New Drugs 20: 311–315. Salvi, S., Fontana, V., Boccardo, S., Merlo, D., Margallo, E., Laurent, S. et al. (2012) Evaluation of CTLA-4 expression and relevance as a novel prognostic factor in patients with non-small cell lung cancer. Cancer Immunol Immunother 61: 1463–1472. Sandler, A., Gray, R., Perry, M., Brahmer, J., Schiller, J., Dowlati, A. et al. (2006) Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 355: 2542–2550. Schreiber, R., Old, L. and Smyth, M. (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331: 1565–1570. Scott, J. et al. (2014) Nivolumab (anti-PD-1; BMS936558, ONO-4538) in combination with platinumbased doublet chemotherapy (PT-DC) in advanced non-small cell lung cancer (NSCLC). J Clin Oncol 32(Suppl.): abstract 8113. Segal, N., Antonia, S.J., Brahmer, J., Maio, M., Blake-Haskins, A., Li, X. et al. (2014) Preliminary data from a multiarm expansion study of MEDI4736, an anti-PD-L1 antibody. J Clin Oncol 32(Suppl.): abstract 3002. Slingluff, C. (2011) The present and future of peptide vaccines for cancer: single or multiple, long or short, alone or in combination? Cancer J 17: 343–350. Spiegel, D., Gettinger, S., Horn, L., Herbst, R.S., Gandhi, L., Gordon, M. et al. (2013) Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC). J Clin Oncol 31(Suppl.): abstract 8008. Steinman, R., Hawiger, D. and Nussenzweig, M. (2003) Tolerogenic dendritic cells. Annu Rev Immunol 21: 685–711.

http://tar.sagepub.com 15

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Therapeutic Advances in Respiratory Disease  Steinman, R., Turley, S., Mellman, I. and Inaba, K. (2000) The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med 191: 411–416. Sterlacci, W., Wolf, D., Savic, S., Hilbe, W., Schmid, T., Jamnig, H. et al. (2012) High transforming growth factor β expression represents an important prognostic parameter for surgically resected non-small cell lung cancer. Hum Pathol 43: 339–349. Sundar, R., Soong, R., Cho, B., Brahmer, J. and Soo, R. (2014) Immunotherapy in the treatment of nonsmall cell lung cancer. Lung Cancer 85: 101–109. Taube, J., Anders, R., Young, G., Xu, H., Sharma, R., McMiller, T. et al. (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4:127ra37. Topalian, S., Hodi, F., Brahmer, J., Gettinger, S., Smith, D., McDermott, D. et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366: 2443–2454. Tumeh, P., Harview, C., Yearley, J., Shintaku, I., Taylor, E., Robert, L. et al. (2014) PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515: 568–571. Vacchelli, E., Aranda, F., Eggermont, A., Galon, J., Sautès-Fridman, C., Cremer, I. et al. (2014) Trial Watch: chemotherapy with immunogenic cell death inducers. Oncoimmunology 3: e27878. Vansteenkiste, J., Cho, B., Vanakesa, T., De Pas, T., Zielinski, M., Kim, M. et al. (2014) MAGRIT, a double-blind, randomized, placebo-controlled Phase III study to assess the efficacy of the recMAGE-A3 + AS15 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive nonsmall cell lung cancer (NSCLC). Ann Oncol 25(Suppl. 4): iv409–iv416. Vansteenkiste, J., Zielinski, M., Linder, A., Dahabreh, J., Gonzalez, E., Malinowski, W. et al. (2013) Adjuvant MAGE-A3 immunotherapy in resected non-small-cell lung cancer: phase II randomized study results. 31: 2396–2403.

Weber, J., Thompson, J., Hamid, O., Minor, D., Amin, A., Ron, I., et al. (2009) A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin Cancer Res 15: 5591–5598. Wesselius, L., Wheaton, D., Manahan-Wahl, L., Sherard, S., Taylor, S. and Abdou, N. (1987) Lymphocyte subsets in lung cancer. Chest 91: 725–729. Whiteside, T. (2010) Immune responses to malignancies. J Allergy Clin Immunol 125(2 Suppl. 2): S272–S283. Wolchok, J., Hoos, A., O’Day, S., Weber, J., Hamid, O., Lebbé, C. et al. (2009) Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 15: 7412–7420. Wolchok, J., Kluger, H., Callahan, M., Postow, M.A., Gordon, R.A., Segal, N. et al. (2013) Safety and clinical activity of nivolumab (anti-PD-1, BMS 936558, ONO-4538) in combination with ipilimumab in patients (pts) with advanced melanoma (MEL). J Clin Oncol 31(15 Suppl.): abstract 9012. Yang, C., Lin, M., Chang, Y., Wu, C. and Yang, P. (2014) Programmed cell death-ligand 1 expression in surgically resected stage I pulmonary adenocarcinoma and its correlation with driver mutations and clinical outcomes. Eur J Cancer 50: 1361–1369. Zheng, H., Li, Y., Wang, X., Zhang, X. and Wang, X. (2010) [Expression and significance of gp96 and immune-related gene CTLA-4, CD8 in lung cancer tissues]. Zhongguo Fei Ai Za Zhi 13: 790–794. Zikos, T., Donnenberg, A., Landreneau, R., Luketich, J. and Donnenberg, V. (2011) Lung T-cell subset composition at the time of surgical resection is a prognostic indicator in non-small cell lung cancer. Cancer Immunol Immunother 60: 819–827. Zou, W. and Chen, L. (2008) Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 8: 467–477.

Vinageras, N., de la Torre, A., Rodríguez, O., Ferrer, C. et al. (2008) Phase II randomized controlled trial of an epidermal growth factor vaccine in advanced nonsmall-cell lung cancer. J Clin Oncol 26: 1452–1458. Visit SAGE journals online http://tar.sagepub.com

SAGE journals

Weber, J., Kähler, K. and Hauschild, A. (2012) Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol 30: 2691–2697.

16 http://tar.sagepub.com

Downloaded from tar.sagepub.com at GEORGIAN COURT UNIV on April 7, 2015

Targeting the immune system to treat lung cancer: rationale and clinical experience.

The use of immunotherapy that harnesses and enhances the innate powers of the immune system to fight cancer cells represents the most promising new ca...
1MB Sizes 0 Downloads 7 Views