Immunotherapeutic Approaches


Mechanism

Specification

1. Reduced antigen processing and presentation

Downregulation or mutation of HLA class molecules

2. Tumour-permissive cytokine profile

Increase of immunosuppressive cytokines: TGF-β, IL-6

Decrease of stimulatory cytokines: IL-2, IFN-γ

3. Immunosuppressive microenvironment

Production of IDO

4. Cellular immune escape

Tregs, M2 macrophages, MDSCs

5. Anergic T cells

5a. by increase of co-inhibitory receptors

5b. by decrease of co-stimulatory receptors

CTLA-4, PD-1, TIM-3, LAG-3

CD137, OX40


HLA human leukocyte antigen, TGFβ transforming growth factor-beta, IL interleukin, IFNγ interferon-gamma, IDO indoleamine 2,3-dioxygenase, Tregs regulatory T cells, MDSC myeloid-derived suppressor cells, CTLA4 cytotoxic T-lymphocyte antigen-4, PD1 programmed death-1, TIM3 T-cell immunoglobulin mucin protein-3, LAG3 lymphocyte-activation gene-3





Immune Checkpoint Inhibitors


Cancer immunotherapy is based on functional restoration of certain signalling pathways of the host immune system that help counteract immune escape of neoplastic tissue. After being recognized as foreign, the tumour can be eliminated. In broad terms, immunotherapy encompasses a variety of diverse treatment approaches like tumour-specific monoclonal antibodies (e.g. cetuximab, bevacizumab), cancer vaccines (e.g. peptide vaccine against HPV, sipuleucel-T), adoptive cell transfer (e.g. autologous tumour-infiltrating lymphocytes, chimeric antigen receptors) and immune-modulating antibodies (e.g. immune checkpoint inhibitors) [24]. The current chapter details the last group, immune checkpoint inhibitors. These drugs, blocking the inhibitory signalling, have brought a fundamental shift in the field of precision medicine, mainly due to their potential to induce durable responses even in patients with refractory disease and cause relatively low incidence of serious adverse events. Tumour-specific antibodies are traditionally excluded from chapters on immunotherapy and are being covered elsewhere, and the remaining two categories are in early phases of clinical testing in head and neck cancer.

By means of receptor-ligand interactions, immune checkpoint pathways regulate the duration and extent of immune system activity to prevent autoimmune reactions. CTLA-4, a receptor expressed on CD4+, CD8+ and regulatory T cells, competitively disrupts the axis between tumour-specific T lymphocytes bearing CD28 receptor and stimulatory ligands CD80 (B7) and CD86 (B70) on antigen-presenting cells. Programmed death-1 (PD-1) is a receptor exposed on the surface of activated T and B lymphocytes and myeloid elements. Programmed death ligand-1 (PD-L1/CD274/B7-H1) and programmed death ligand-2 (PD-L2/CD273/B7-DC) are transmembrane proteins found on a wide variety of cells transmitting a negative signal downregulating T-lymphocyte activation. An impaired immune recognition may thus occur in case of a high fraction of CTLA-4 or PD-1 positive T cells in the tumour microenvironment or an increased expression of PD-L1 or PD-L2 by the tumour itself. Importantly, while PD-1 is a receptor for both PD-L1 and PD-L2 ligands, PD-L1 binds also to B7-1 receptor located on T cells. Consequently, although PD-1 blockage interferes with interactions between PD-1 and its two ligands, PD-L1 can still activate B7-1 receptor [23, 25].

During the application of immune checkpoint inhibitors, practitioners may encounter two specific situations concerning the distinctive kinetics of antitumour response and the characteristic, immune-related side effects. First, pseudoprogression, rare in SCCHN patients, describes a transient clinical phenomenon detectable early after treatment onset. Although resembling true neoplastic growth, it merely reflects inflammatory changes generated by immune cell infiltration. In addition, compared to classic cytotoxic agents, immunotherapy often elicits delayed responses as it may take some time to unlock the natural anticancer potential of the immune system. Consequently, this modality may not be appropriate for patients with rapidly progressive, symptomatic disease, who require prompt relief. Moreover, such a unique behaviour of cancer upon exposure to immunotherapy warrants specific immune-related response criteria, since traditional measurement guidelines do not allow for treatment beyond initial progression [26, 27]. Based on survival analysis of 592 melanoma patients treated with pembrolizumab, Hodi et al. showed that conventional Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST v1.1) might underestimate the benefit of pembrolizumab in about 15 % of cases [28].

Secondly, autoimmune reactions affect a significant proportion of patients and appear usually 6–12 weeks following therapy initiation. The most frequent are endocrinopathies (hypophysitis, thyroiditis, adrenal insufficiency), diarrhoea, colitis and maculopapular rash, but they also include general symptoms like fevers, chills, and lethargy, hepatotoxic side effects, neuropathies and less commonly pneumonitis [29]. In a phase II trial exploring ipilimumab among 155 subjects with pretreated melanoma, immune-related adverse events were reported in 70 % of study population with grade 3–4 toxicities observed in 34 (22 %) patients [30]. To prevent unnecessary treatment discontinuation or enable an early re-initiation, timely diagnosis and appropriate management are warranted. Glucocorticoids can reverse nearly all immune-related adverse reactions but should be employed only for grade 3–4 or prolonged grade 2 toxicities [29].

There are several immune checkpoint inhibitors undergoing clinical evaluation in head and neck cancer. Two monoclonal antibodies of the immunoglobulin (Ig) G4 type with a high affinity for PD-1, pembrolizumab (MK-3475) and nivolumab (ONO-4538), have already been approved for treatment of advanced melanoma and lung cancer. Durvalumab (MEDI4736), atezolizumab (MPDL3280A) and avelumab (MSB0010718C) belong to an evolving category of monoclonal antibodies against PD-L1. As indicated above, ipilimumab (MDX-010) entered the market as a practice-changing drug for melanoma patients. A phase Ib study exploring ipilimumab with cetuximab and irradiation is ongoing in patients with locally advanced head and neck cancer (NCT01860430), whereas tremelimumab, another CTLA-4-directed antibody, is being studied in the recurrent/metastatic setting (see below).


Immunotherapy in R/M-SCCHN


SCCHN is a suitable candidate for the immunotherapeutic approach. As an immunosuppressive disease, SCCHN utilizes several mechanisms to evade immunosurveillance. It manipulates its own immunogenicity, produces immunosuppressive mediators (e.g., transforming growth factor-beta, interleukins 6 and 10) and promotes immunomodulatory cell types (myeloid-derived suppressor cells, suppressive regulatory T cells, tumour-associated macrophages) [23]. Immune dysfunction has been implicated in carcinogenesis of HPV-related oropharyngeal cancer as well as the majority of remaining SCCHN cases which are linked to smoking and alcohol. Oncogenic types of HPV, such as 16 and 18, encode E6 and E7 proteins which, together with host and environmental factors, contribute to the ability of the virus to persist in the infected organism for long periods of time. Tobacco-driven tumours harbour chronic inflammation and a high mutational load leading to increased immunogenicity. In this regard, establishment of T-cell tolerance to chronic HPV infection and overexpressed or mutated antigens poses one of the key mechanisms of immune escape [23, 31]. Recently, using gene expression-based consensus clustering, copy number profiling and HPV status, five new subtypes of SCCHN were identified to overcome some of the limitations associated with traditional classification based on anatomic site and stage. Two of these subtypes, one HPV-positive and one negative, demonstrated a prominent immune and mesenchymal phenotype with marked CD8+ lymphocyte infiltration. Such a strong activation of the immune system provides a further rationale for immunotherapy and may become a predictive biomarker for this therapeutic approach [32].

As outlined above, head and neck cancer ranks among cancers with higher somatic mutation rates. In lung cancer, a high mutation frequency was attributed to smoking and implicates increased immunogenicity due to an increment in tumour neoantigens [33]. Interestingly, the efficacy of anti-PD-1 and anti-PD-L1 antibodies seems to correlate with tobacco exposure [3436]. This finding is intriguing within the context of head and neck cancer considering its close etiological relationship to tobacco abuse. Moreover, a subset analysis of 42 patients with melanoma, non-small-cell lung, colorectal, renal-cell and prostate cancers treated with an anti-PD-1 antibody indicated an association between PD-L1 expression on tumour cells and objective response [37]. In head and neck cancer including nasopharyngeal carcinoma, several reports confirmed that PD-L1 expression is a common event found in 46–100 % of tumour samples [38, 39]. However, these data should be used with caution because of variable quality of archival tissue specimens and differences in the used assays and scoring methods. Nonetheless, they add further evidence to the prominent immune phenotype of head and neck cancer.

The majority of R/M-SCCHN patients qualify for palliative measures with an expected survival of 6–10 months. At present, according to the pivotal EXTREME (Erbitux in first-line treatment of recurrent or metastatic head and neck cancer) trial, the platinum/5-fluorouracil/cetuximab regimen is the approved first-line systemic treatment for fit patients [40]. Unfortunately, the long-term follow-up data are far from being satisfactory underscoring the burning need for further improvement [41]. The following part of this chapter describes the preliminary results from two early clinical trials exploring pembrolizumab and durvalumab as well as the recently presented, possibly practice-changing phase III data of nivolumab.


Pembrolizumab


KEYNOTE-012 is an ongoing non-randomized, multi-cohort phase Ib trial (NCT01848834), which recruited participants with a diagnosis of SCCHN, bladder, triple-negative breast and gastric cancers. At the 2014 American Society of Clinical Oncology (ASCO) annual meeting, the first results of cohort B from the KEYNOTE-012 study were made public [42]. Cohort B consisted of 56 PD-L1 positive R/M-SCCHN cases with or without previous systemic therapy. Pembrolizumab at a dose of 10 mg/kg intravenously every 2 weeks yielded a 20 % overall response rate. Subgroup analysis based on HPV status found comparable overall response rates, while median progression-free and overall survivals were longer in HPV-positive than negative patients (17.2 vs. 8.1 weeks and median overall survival not reached vs. 9.5 months, respectively). The most frequently observed toxicities were fatigue (18 %), pruritus (10 %) and nausea (8 %) [42, 43].

At ASCO 2015, results from cohort B2 of this study were released. Patients with R/M-SCCHN, regardless of PD-L1 expression, HPV status or prior systemic therapy, were eligible for inclusion if they had measurable disease based on RECIST v1.1 and Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. Pembrolizumab was given at a fixed dose of 200 mg intravenously every 3 weeks until either progression, intolerable toxicity, or a 24-month treatment limit was reached. The investigators permitted treatment beyond progression as well as pembrolizumab rechallenge. Tumour assessments were performed at baseline and every 8 weeks with radiographic imaging. Primary objectives of this trial are overall response rate (ORR) per investigator evaluation (RECIST v1.1) and adverse events determined according to the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Secondary end points include progression-free survival, overall survival and duration of response.

As of March 2015, 132 predominantly male patients with a median age of 60 years (range, 25–84) were enrolled. Twenty-two percent had received one and almost 60 % two or more prior lines of therapy for R/M disease. Overall, pembrolizumab was well tolerated; no treatment-related deaths occurred. Drug-related adverse events of any grade were reported in 79 subjects (60 %) with the most frequent one being fatigue (15 %) followed by hypothyroidism (9 %). Other side effects developing at a rate of at least 5 % comprised decreased appetite (8 %), rash (8 %), dry skin (7 %), pyrexia (7 %), arthralgia (5 %), nausea (5 %) and weight loss (5 %). Thirteen patients (10 %) experienced severe acute toxicity (grade 3–4), most commonly swelling of the face and pneumonitis observed in two cases each. Concerning adverse events of special interest, which are those potentially associated with the investigational compound, they included the following toxicity, apart from the already mentioned hypothyroidism and pneumonitis: grade 1–2 thyroiditis (2 %), grade 3 colitis (1 %), grade 1–2 interstitial lung disease (1 %), grade 1–2 acquired epidermolysis bullosa (1 %), grade 3 drug-induced liver injury (1 %), grade 1–2 epidermolysis (1 %) and grade 4 diabetic ketoacidosis (1 %). Patients with pneumonitis, colitis, interstitial lung disease and drug-induced liver injury required treatment discontinuation.

Efficacy analyses were performed in 117 subjects who received one or more doses of pembrolizumab, had measurable disease at baseline and at least one post-baseline scan or discontinued therapy due to progressive disease or drug-related adverse events. Pembrolizumab produced overall response, complete response, partial response and disease control rates of 25 %, 1 %, 24 %, and 50 %, respectively. More than half of the study group (56 %) experienced tumour shrinkage of any degree. Both HPV-positive and negative cases benefited from the drug with a slightly higher response rate in the latter group (21 % versus 27 %). Interesting results were found in the exposure-response analysis. Responses occurred largely at 8 or 16 weeks corresponding with the time of imaging; median time to response was 9 weeks. Two patients had a late response at about 25 weeks. Median time of follow-up was 6 months, but it was not reached for the duration of response. Importantly, 40 patients including many of those with stable disease continued receiving pembrolizumab. Moreover, at the time of data cut-off, 86 % (25/29) of the responding patients had an ongoing response [27].

Subsequently, a pooled analysis of the two R/M-SCCHN cohorts from the KEYNOTE-012 trial (a total of 173 patients, 56 from cohort B and 117 from cohort B2) were reported. The aim of this post hoc analysis was to identify R/M-SCCHN subgroups that may derive the greatest clinical benefit from pembrolizumab. The anti-PD-1 antibody achieved an overall response rate of 24 % with two cases of complete remission. The response rate was higher in patients who received two or less previous treatment lines versus those with more prior therapies (32 % versus 14 %). Also, response rates were higher in patients with smaller tumour sizes: 36 % in those in whom the tumour size was ≤ median value versus 15 % in those with larger tumours. In this respect, HPV status did not confer any advantage. Median progression-free and overall survival for the whole population (n = 173) were 2.2 (95 % confidence interval [CI], 2.0–3.6) and 9.6 months (95 % CI, 6.6-not reached), respectively. Median follow-up time was 13.7 and 5.7 months in cohorts B and B2, respectively [44]. When compared with results achieved by various epidermal growth factor receptor (EGFR) inhibitors in second-line therapy, progression-free survival appears not to be much different as opposed to response rate and overall survival clearly favouring pembrolizumab [4450] (Table 15.2). Even more convincing data are available for the randomized trial with nivolumab as described below.


Table 15.2
Trials with PD-1-directed agents and randomized studies of EGFR inhibitors in second-line treatment of R/M-SCCHN




























































Study, author (year)

N

Regimen (treatment arms A, B, C)

Response rate (%)

Median PFS (months)

Median OS (months)

KEYNOTE-012 (cohort B and B2)

Chow (2015) [44]

173a

Pembrolizumab

23.7

2.2

9.6

CHECKMATE-141

Gillison (2016), Ferris (2016) [45, 46]

361

A: Nivolumab

B: MTX or D or cetuximab

22.2b, c

2.3b

3.2b, c

2.0b

7.5c

5.1

IMEX

Stewart (2009) [47]

486

A: Gefitinib (250 mg)

B: Gefitinib (500 mg)

C: MTX

2.7

7.6

3.9

ND

ND

ND

5.6

6.0

6.7

ECOG 1302

Argiris (2013) [48]

270

A: D + Gefitinib

B: D + placebo

12.5

6.2

3.5 (TTP)

2.1 (TTP)

7.3

6.0

ZALUTE

Machiels (2011) [49]

286

A: Z + BSC

B: BSC (optional MTX)

6.3

1.1

2.3c

1.9

6.7

5.2

LUX-Head & Neck 1

Machiels (2015) [50]

483

A: Afatinib

B: MTX

10.2

5.6

2.6c

1.7

6.8

6.0


PD1 programmed death-1, EGFR epidermal growth factor receptor, N number of randomized patients, MTX methotrexate, D docetaxel, Z zalutumumab, BSC best supportive care, PFS progression-free survival, ND no data, TTP time to progression, OS overall survival

aEvaluable patients

bFor programmed death ligand-1 (PD-L1) expression ≥5 %

cSignificant differences

An exploratory biomarker analysis confirmed that PD-L1 expression by immunohistochemistry correlates with response. However, pembrolizumab exhibits activity also in PD-L1 negative cases. Using a preliminary PD-L1 cut-point derived from the Youden index, it could be demonstrated that tumours with a value above this cut-point responded better (46 %, 5/11) than those underneath (11 %, 5/44) [42]. Very recently, Yearley et al. shed more light on this issue introducing a novel immunohistochemistry assay for PD-L2, which was verified also on head and neck cancer samples. As a result, the authors stated that PD-L2 expression may be present even in the absence of PD-L1, which partly explains the phenomenon of PD-L1 negative patients responding to PD-1-directed agents. In this regard, patients resistant to anti-PD-L1 antibodies might still have benefit from anti-PD-1 drugs blocking both PD-L1 and PD-L2 signal transduction pathways [51]. Another area of biomarker research concerns gene-expression signatures. Using NanoString technology, Seiwert and colleagues tested 43 PD-L1 positive patients enrolled in the KEYNOTE-012 trial. The investigators proved a statistically significant correlation between progression-free survival and inflamed phenotype consisting of four previously established multigene expression signatures (interferon-γ, T-cell-receptor signalling, expanded-immune, and de novo). Furthermore, interferon-γ signature was significantly associated with best overall response and, interestingly, it has been linked to the independently discovered inflamed/mesenchymal SCCHN subtype alluded to above [52].


Nivolumab


Another PD-1-directed agent, nivolumab, has recently been brought to the spotlight of healthcare professionals as the first drug ever to show a survival benefit in patients with platinum-refractory R/M-SCCHN. At American Association for Cancer Research (AACR) annual meeting in New Orleans in April 2016, the investigators of the randomized, global CHECKMATE-141 phase III trial (NCT02105636) announced that the study was stopped early, after a planned interim analysis performed by an Independent Data Monitoring Committee, because the statistical boundary for overall survival was crossed. The trial evaluated the efficacy and safety of nivolumab at a dose of 3 mg/kg intravenously every two weeks versus weekly intravenous single-agent chemotherapy (methotrexate 40 mg/m2, docetaxel 30 mg/m2) or cetuximab (400 mg/m2 once, then 250 mg/m2) in a 2:1 ratio. Key eligibility criteria were as follows: R/M-SCCHN of the oral cavity, pharynx or larynx not amenable to curative therapy, progression on or within 6 months of platinum-based chemotherapy, good ECOG performance status (0 or 1), no active brain metastases and p16 immunohistochemistry to determine HPV status. Prior cetuximab treatment served as a stratification factor. Primary endpoint was overall survival and secondary endpoints included progression-free survival, overall response rate, safety, duration of response, biomarkers and quality of life.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 9, 2017 | Posted by in ONCOLOGY | Comments Off on Immunotherapeutic Approaches

Full access? Get Clinical Tree

Get Clinical Tree app for offline access