Skip to main content
Immunology logoLink to Immunology
. 2015 Mar 30;145(2):182–201. doi: 10.1111/imm.12459

New clinical advances in immunotherapy for the treatment of solid tumours

Valentina A Zavala 1, Alexis M Kalergis 2,3,4,5,
PMCID: PMC4427384  PMID: 25826229

Abstract

Advances in understanding the mechanisms of cancer cells for evading the immune system surveillance, including how the immune system modulates the phenotype of tumours, have allowed the development of new therapies that benefit from this complex cellular network to specifically target and destroy cancer cells. Immunotherapy researchers have mainly focused on the discovery of tumour antigens that could confer specificity to immune cells to detect and destroy cancer cells, as well as on the mechanisms leading to an improved activation of effector immune cells. The Food and Drug Administration approval in 2010 of ipilumumab for melanoma treatment and of pembrolizumab in 2014, monoclonal antibodies against T-lymphocyte-associated antigen 4 and programmed cell death 1, respectively, are encouraging examples of how research in this area can successfully translate into clinical use with promising results. Currently, several ongoing clinical trials are in progress testing new anti-cancer therapies based on the enhancement of immune cell activity against tumour antigens. Here we discuss the general concepts related to immunotherapy and the recent application to the treatment of cancer with positive results that support their consideration of clinical application to patients.

Keywords: cancer, clinical, immunotherapy, programmed cell death 1, T-lymphocyte-associated antigen 4, trial

Introduction

By the mid-twentieth century, the development of chemotherapy as an adjuvant to radiation therapy and surgery was a significant advance for leukaemia and lymphoma treatment. Later, these approaches impacted the treatment of solid tumours.1 Nevertheless, chemotherapy, as a systemic unspecific treatment, also affects non-malignant cells and contributes to generalized adverse secondary effects. The search for specific targets to treat cancer developed slowly, but the discovery of specific drugs, such as imatinib by the end of the 1990s, a tyrosine-kinase inhibitor now approved by the The Food and Drug Administration (FDA) for the treatment of several types of cancers, made chemotherapy an efficient targeted therapy. Along these lines, the use of antibodies to target specific proteins on the membrane of tumour cells became especially relevant in cancer treatment and, for the first time in 1997 the FDA approved rituximab for the treatment of B-cell lymphomas. These results led to the subsequent approval of other antibodies, such as trastuzumab and bevacizumab for breast and colon cancer treatment, respectively. Furthermore, the increasing evidence that the cancer microenvironment, composed mainly of extracellular matrix components, fibroblasts, immune cells, among other normal cells, could modulate cancer cell phenotype and proliferation through paracrine and yuxtacrine signalling2 led to the notion that these components can control tumour development. Indeed, the fact that immune cells are present as lymphocyte infiltrates in several solid tumours, such as breast, prostate, pancreatic and colon cancer, has revealed the active role of humoral and cellular immune response on tumour suppression.37 Leucocyte infiltrates are composed of cells of lymphoid and myeloid origin, mainly dendritic cells, macrophages, natural killer cells, granulocytes and mast cells, which interact directly and indirectly with tumour and stromal cells to modulate the tumour microenvironment.3 During the first steps of cancer progression, immune cells promote an inflammatory response aimed to repair tissue damage through the secretion of growth factors, interleukins, chemokines, histamine and other bioactive molecules.2,3,8 Nevertheless, the chronic exposure of tumour cells to these molecules can eventually promote angiogenesis, proliferation and tumour development.2 Further, the specific composition of infiltrating immune cells has been correlated with tumour progression, regression and treatment response5,6,9 and the quantification of infiltrating immune cells can also be used as a prognostic factor, known as ‘immunoscore’.10

According to the National Cancer Institute definition, immunotherapy is any ‘treatment to boost or restore the ability of the immune system to fight cancer, infections, and other diseases’.11 Under this concept, cytokines are master immunomodulators because they have autocrine and paracrine function, being able to promote or inhibit cellular and humoral responses. In 1998, the use of high-dose interleukin-2 (IL-2) was approved by the FDA to treat melanoma, which was a significant advance in cancer treatment, because by targeting the immune system it was possible to successfully control the disease.12 However, cytokines alone are not enough to elicit a complete and efficient immune response for cancer treatment, and so far, together with the use of cytokines, monoclonal antibodies and cell-derived vaccines are the most developed approaches for this purpose. Several of these approaches have been evaluated in several clinical trials, confirming high efficiency and specificity in cancer treatment, and in consequence, have been approved by the FDA for cancer treatment.1316 Here we resume the basic approaches for immunotherapy development and review some of the most successful clinical trials of the last 4 years for the most common and studied types of solid cancers.

Antigenic targets for immunotherapy

The development of new immunotherapeutic approaches for cancer treatment has developed along with the discovery of new tumour antigens as immunogenic targets.17 On the one hand, there are tumour antigens that play an essential role in tumour growth or progression and that can be targeted by cytotoxic cells and antibodies.8 These antigens are called oncoantigens8 and can be divided into three main classes according to their cellular localization. Class I antigens are exposed on the membrane of tumour cell proteins that are over-expressed compared with normal cells, such as the over-expression of human epidermal growth factor receptor 2 (HER2) in breast and gastric cancer, among others. Class II antigens are proteins and other molecules secreted by tumours or environmental molecules, of which certain concentrations facilitate tumour development. Finally, Class III antigens, are intracellular antigens that cannot be recognized by immune cells because they present intracellular localization,18 especially for those proteins localized in the nucleus.8

In addition, there are some tumour-associated antigens (TAAs) that cannot be classified as oncoantigens because they do not necessarily promote tumour development, but rather they can be efficiently used as tumour biomarkers and/or to improve the specific anti-tumour immune reaction. They may be the product of a somatic mutation that causes a truncated protein that is recognized by the antigen-processing machinery and presented on the major histocompatibility complex class I (MHC-I) molecules by tumour cells. Also, oncofetal proteins, normally expressed during fetal development, can be found aberrantly expressed during adulthood, such as the extensively studied α-fetoprotein as well as carcinoembryonic antigen.19 In the case of secreted antigens, Spondin-2, a secreted extracellular matrix protein has been identified as a reliable diagnostic biomarker for prostate cancer and to predict chemotherapy responses.20 Carcinoembryonic antigen can also be found in serum of other cancer patients, mainly in rectal cancer and non-small-cell lung cancer (NSCLC) patients, is used as a biomarker for chemotherapy response and prognosis.2123 Also, we would add exosomes to this class, which are an interesting target for immunotherapy because they express MHC-II and co-stimulatory molecules that can eventually activate tumour antigen-specific T cells. However, it has been also shown in a tumour-bearing mouse model that tumour-derived exosomes can inhibit the anti-tumour immune response.24 Breast cancer and B-cell lymphoma patients can show resistance to monoclonal antibody therapy due the secretion of exosomes containing HER2 and CD20, which sequester trastuzumab and rituximab, respectively, the therapeutic antibodies used for the treatment of these cancers.25,26 To overcome this problem, a therapeutic haemofiltration approach has been suggested to selectively capture and remove exosomes from the circulatory system.27 In the case of intracellular antigens, new strategies have focused on the liberation of intracellular antigens from the cells to induce a specific immune response. For example, Noguchi et al. showed in a mouse model that combined treatment of chemotherapy plus a targeted monoclonal antibody against Cancer-testis antigen (NY-ESO-1), an intracellular protein expressed by cancer cells but not normal cells (except in testis germ cells28), can induce an efficient CD8+ T-cell response.29 Chemotherapy drugs, which induce the release of NY-ESO-1 from necrotic cells, allow anti-NY-ESO-1 antibodies to form in situ immune complexes and induce a higher number of effector/memory NY-ESO-1-specific CD8+ T cells that secrete more interferon-γ and/or tumour necrosis factor-α compared with drug treatment alone. These combined therapy-treated mice showed an improved survival due to a long-lasting anti-tumour capacity.29

It is important to point out that most of these antigens are normally expressed by cells in healthy tissues. One of the major challenges for the development of cancer vaccines is the determination of the optimal and most specific tumour antigens to be used as target for immunotherapy. According to this notion, a good antigen should display high antigenicity and a homologous expression in tumour tissue to overcome the escape of tumour cells due to tumour heterogeneity. On the basis of this knowledge, a translational research project carried out by the National Cancer Institute focused on the discovery of new and common tumour antigens among different cancers.30 For this purpose, they selected 75 candidate antigens for further analyses using different approaches to determine their potential use in the development of new anti-cancer vaccines. Even though none of the 75 antigens had all the expected characteristics for a cancer antigen (listed in ref. 30), 46 of them were immunogenic based on clinical trials and 20 were suggested to be potential targets for immunotherapy. Moreover, there are other approaches being used for the discovery of new tumour antigens as potential immunotherapy targets. For example, Walter et al.31 used an antigen discovery platform called XPRESIDENT for the development of a new vaccine for the treatment of renal-cell cancer (RCC). This platform allowed the identification, selection and validation of multiple tumour-associated peptides naturally expressed in clinical cases and is based on genomics, proteomics and bioinformatics approaches and on T-cell immunology. Also, ImmunoCore, a biotechnology company located in Abingdon, UK (http://www.immunocore.com/contact/), has focused in the search for specific T-cell targets for the development of cancer therapies. So far, they have created a bi-specific drug called IMCgp100, a soluble, affinity enhanced T-cell receptor specific for a fragment of gp100, a melanoma-specific antigen, fused to an anti-CD3 specific antibody single-chain variable fragment, which confines killer T cells to where melanoma cells are and induces their destruction.32 A phase I/II clinical trial is currently in progress to assess the safety profile and to establish a tolerable dose of IMCgp100 in patients with HLA-A2-positive malignant melanoma (NCT01211262).33,34

Immunotherapy types

Active therapy comprises the destruction of tumour cells by a direct effect or by indirectly stimulating immune responses.35 One of the most frequently used strategies is to take advantage of soluble molecules, such as cytokines,8 which are independent of antigen recognition by host immune cells. Such an approach is known as non-specific-antigen immunotherapy. Interleukin-2 and interferon-α have been approved by the FDA, but there are others soluble molecules, such as IL-7 and granulocyte–macrophage colony-stimulating factor (GM-CSF), that are being used in human trials to enhance anti-cancer therapy (Table1). Also, the allogeneic bone marrow transplant is a recurrent alternative passive therapy for haematological and some solid cancers. Although allogeneic transplant of peripheral blood stem cells has shown better results, it has been observed that the main advantage of bone marrow transplant is a reduced probability of relapsing in some haematological cancers.36

Table 1.

Types of immunotherapy against cancer

Antigen-specific Antigen-non-specific
Passive Adoptive cell transfer:
  • LAK cells, TILs + IL-2

  • Monoclonal antibodies:

  • Anti-HER2, anti-EGFR, anti-VEGF

Bone marrow transplantation
Active Cancer vaccines:
  • Tumour antigen-based vaccines

  • Peptide-based vaccines

  • DC-based vaccines

  • Vector-based vaccines

  • Idiotype-based vaccines

Cytokines: IL-2, IL-7, IL-12, IFN-γ, IFN-α, GM-CSF, etc.
  • Immunomodulatory monoclonal antibodies: Anti-PD-1, anti-CTLA-4, anti-CD20, etc.

Abbreviations: DC, dendritic cell; GM-CSF, granulocyte–macrophage colony-stimulating factor; IFN, interferon; IL-2, interleukin-2; LAKs, lymphokine-activated killer; TILs, tumour infiltrating lymphocytes.

Immune molecular checkpoints, which work as main regulators of the tolerance/immunity balance, have become a useful target for immunotherapy across different types of cancer because of the independence of antigen-specific immune reactions. Along these lines, checkpoint receptor inhibitors, such as anti-T-lymphocyte-associated antigen 4 (CTLA-4) and anti-Programmed Cell Death 1 (PD-1) antibodies have demonstrated a high efficacy for the treatment of patients with different types of tumours.37 PD-1 is a co-receptor expressed on peripheral CD4+ and CD8+ T cells, and the co-stimulatory pathway activated by their ligands, Programmed death-ligand 1 (PD-L1) (B7-H1) and PD-L2 (B7-DC) expressed in peripheral tissues, such as tumour or stromal cells, deliver inhibitory signals that block T-cell proliferation and induce immunological tolerance. PD-L1 is mostly expressed on haematopoietic, non-haematopoietic cells and also tumour cells, in contrast to PD-L2, which is normally restricted to fewer cell types.38 A Phase I clinical trial (NCT00730639) has tested an anti-PD-1 monoclonal antibody called BMS-936558 (also known as MDX-1106 or ONO-4538) in patients with advanced melanoma, RCC, NSCLC, colorectal cancer or castration-resistant prostate cancer.37 Although the disease progression was the most common cause of death, there was more extensive survival in patients with NSCLC, melanoma, or RCC treated with the antibody in comparison to those treated with chemotherapy and/or kinase inhibitors.37 As PD-L1 expression was associated with an objective response to treatment, the authors suggested that its expression might be used as a molecular biomarker to identify those patients that could be treated with anti-PD-1 antibody. Currently, there are two ongoing clinical trials (in phase I and phase II) testing the pharmacodynamic and biological properties of anti-PD-1 antibodies in patients with RCC (NCT01358721 and NCT01354431). In addition to these studies, another phase I clinical trial has been published (NCT00729664), in which an anti-PD-L1 antibody was tested in RCC, NSCLC, melanoma, gastric cancer, ovarian cancer, colorectal cancer, pancreatic cancer and breast cancer.39 An objective response reflected in a durable tumour regression of 6–17% was observed, and a prolonged disease stabilization in patients with metastatic NSCLC, melanoma, RCC or ovarian cancer.39 As NSCLC is considered non-immunogenic and non-responsive to immunotherapies, the 10% of objective response obtained in these patients is remarkably significant and suggests this pathway as a target for therapeutic intervention for this cancer and other related cancers.39 In both studies, neither colon cancer nor pancreatic cancer patients had tumour responses after anti-PD-1 or anti-PD-L1 antibody treatment. The use of anti-PD-1 ligand antibody is a promising approach because inhibitory ligands are found to be expressed in tumour cells in response to the immune system activation, as part of a mechanism called adaptive resistance.40 Regarding anti-PD-L1 treatment, two phase I clinical trials (NCT0137584241 and NCT0137584242) were recently published, in which a monoclonal antibody against PD-L1 (MPDL3280A), was evaluated for safety and tolerability in metastatic urothelial bladder cancer and various cancer types. To test whether PD-L1-positive patients might specifically respond to MPDL3280A, patients were selected based on the expression of PD-L1 in tumour-infiltrating immune cells, which was evaluated by immunohistochemistry. An objective response rate (ORR) equal to 43% was obtained for PD-L1-positive previously treated metastatic urothelial bladder cancer patients after 6 weeks of follow up, 7% of them had a complete response rate. After 12 weeks of follow up, ORR was 52%.41 Complete and partial response was 18% among all tumour types,42 with progression-free survival (PFS) of 18 weeks. Taken together these data suggest that blockade of the interaction between PD-1 and its ligands could significantly contribute to increase the efficiency of tumour immunity induced by other treatments. An extensive review of PD-1 and PD-L1 has been recently published elsewhere.43

Importantly, additional checkpoint molecules, such as T-cell immunoglobulin domain and mucin domain-3 (TIM-3) and lymphocyte activation gene 3 (LAG3), have been found over-expressed in cells4446 and their potential use in immunotherapy has been reported in pre-clinical studies.4749 Special attention has been paid to LAG-3, which is up-regulated by T cells exhausted by chronic antigen exposure and especially over-expressed in tumour-infiltrating lymphocytes (TILs).45,46 LAG-3 and PD-1, are co-expressed by TILs,45 and display synergistic functions at controlling immunological homeostasis. It has been recently shown that dual inhibition of both receptors decreased tumour growth and enhanced anti-tumour immunity.50 Because dual treatment promotes tumour-specific immune responses, with reduced unspecific or self-antigen-specific responses, it can be considered as an interesting strategy for further evaluation in the clinic. Along these lines, a phase I clinical trial is in progress to assess the safety and tolerability of the anti-LAG-3, BMS-986016, alone and in combination with anti-PD-1 in subjects with advanced solid tumours (NCT01968109).

The main aim when designing a new immune approach to treat cancer is not only to specifically target an antigen to direct the immune response against tumour cells only, but also to enhance the natural immune response against it, together with creating immunological memory to obtain a prolonged anti-tumour response. However, this aim becomes difficult because during tumour development, tumour cells might lose antigens for which an immune response was established and also express new tumour-specific antigens because mutations may occur in this process. Hence, antigen-specific immunotherapy requires a well-defined and stable antigen target that can elicit an efficient immune response. Passive, antigen-specific immunotherapies involve the use of the immune system components that are already able to promote an immune response against a specific antigen. Cell-based and antibody-based approaches have been developed for this purpose. The adoptive cell transfer basically comprises the transfer of TILs to patients because there is wide evidence that these immune cells participate in tumour regression in different solid tumours.51 TILs are obtained from tumours and are cultured with IL-2 to promote the expansion of the tumour antigen-specific T cells. After 6 or 8 weeks of culture, the increased number of cells can be transferred into the host where TILs localize to the tumour and start an in situ anti-tumour reaction.35The use of antibodies generated ex vivo against a particular antigen works also as an adoptive transfer of immunity that generates a specific humoral response. This approach has been widely used for the treatment of diverse types of cancer as will be discussed below. The active specific treatments require host immune-competence and involve the use of vaccines. Several types of vaccines are being developed, from purified antigen vaccines to polyvalent antigen vaccines, depending on the immunotherapy purpose. The main challenge is to generate an immune response to a variety of tumour antigens at the same time. Briefly, the main vaccine subtypes developed so far are (i) tumour antigen-based, (ii) peptide-based, (iii) dendritic cell-based, (iv) vector-based and (v) idiotype-based vaccines (for an exhaustive revision see ref. 35). Hobo et al.,52 developed a dendritic cell (DC)-based vaccine with improved immunogenic potential by transfecting DCs with the small-interfering RNAs (siRNAs) of PD-1 ligands combined with a target antigen mRNA electroporation. They could specifically silence PD-L1 and PD-L2 with high efficiency and keep the phenotype and migratory capacity of DCs using a lipid nanoparticle whose cationic lipids efficiently mediate siRNA delivery into the cells. Besides, they electroporated DCs with allogeneic minor histocompatibility antigen mRNA and demonstrated a superior stimulatory potential and an effective ex vivo boost of antigen-specific CD8+ effector/memory T-cell responses in transplanted cancer patients. Although T cells over-express PD-1 during T-cell activation, expanded antigen-specific T cells would not be down-modulated because DCs no longer express PD-1 ligands, keeping T cells in a highly activated state. These results allowed authors to suggest that siRNA-modified DCs can be clinically used for boosting the immune response in transplanted cancer patients to induce T-cell-mediated anti-tumour immunity without evoking a response against healthy host cells. Another group has also developed the use of mRNA-based modification of DCs to induce a specific T-cell response against tumour cells whose TAAs have not yet been identified. Yu et al.,53 transfected autologous DCs from osteosarcoma tumour-bearing rats with allogeneic mRNA from osteosarcoma cell lines. Vaccinated rats showed a longer survival due to the induction of a specific long-term T-cell memory immune response.

Because each cancer develops its own unique antigenic profile as the result of the histological origin and the interaction of tumour cells with immune cells and the microenvironment, new anti-cancer strategies developed so far for most common cancer types will be discussed individually in the next sections (Table2). Because it is difficult to review all the immunotherapeutic approaches that are currently being developed for various types of cancer, we will focus on clinical trials that have shown promising results for the most common types of solid tumours.

Table 2.

Selected cancer immunotherapy clinical trials

Clinical trial Reference Phase Treatment regimen Patients Response Adverse events Type
Melanoma
III Ipilimumab 3 mg/kg per 3 weeks up to 4 treatments
  • Ipilimumab + gp100 Ipilimumab alone

  • Gp100 alone

676
HLA*0201 + unresectable stage III or IV melanoma, previously treated with chemotherapy and/or temozolomide with IL-2 doses OS: 10 and 6·4 month
  • ipilimumab + gp100 versus gp100 alone, respectively

  • HR: 0·68, P < 0·001

  • OS: 10·1 ipilimumab alone

  • HR: 0·66, P: 0·003

Grade 3 or 4 irAE in 10–15% of patients with ipilimumab and 3% gp100 alone. 7 deaths related to irAEs Monoclonal anti-CTLA
III Ipilimumab 10 mg/kg + DTIC
  • Placebo + DTIC

  • Given at week 1, 4, 7, 10 followed by DTIC alone every 3 weeks until week 22

502 untreated metastatic melanoma stage IIIC or IV patients OS: 11·2 month ipilimumab + DTIC OS: 9·1 month DTIC + Placebo Higher SR at 1 year (47·3% versus 36·3%) and 2 year (28·5% versus 17·9%) 3 year (20·8% versus 12·2%), respectively HR: 0·72, P < 0·001 Grade 3 or 4 AEs in 56·3% of ipilimumab + DTIC versus 27·5% DTIC + placebo (P < 0·001) No drug related deaths Monoclonal anti-CTLA
III Nivolumab injections every 2 weeks for up to 96 weeks 107 advance melanoma patients OS: 16·8 month 1 and 2 year SR for nivolumab versus control were 62% and 43%, respectively PFS: 3·7 month 1, 2 year PFS rates: 36% and 27%, respectively Objective responses in 31% of treated patients Most of AEs within the first 6 months of therapy Grade 3 or 4 pneumonitis (1%), fatigue (32%), rash (23%), diarrhoea (18%) 22% grade 3 or 4 presented treatment-related AEs Monoclonal Anti-PD-1
II Non-randomized cohort of ipilimumab-treated (IPI-T) and ipilimumab-naive (IPI-N) treated with different doses of pembrolizumab A randomized cohort was also treated 411 melanoma IPI-T or IPI-N patients Durable responses (88% by October, 2013) ORR: 40% IPI-N and 28% in IPI-T PFS: 24 versus 23 weeks, respectively OS not reached (1 year OS: 71%) 12% of patients experienced drug-related grade 3–4 AEs, 4% discontinued due drug-related AEs No drug-related deaths Monoclonal Anti-PD-1
  Hamid et al.70 I Intravenous pembrolizumab 10 mg/kg every 2 or 3 weeks or 2 mg/kg every 3 weeks 135 patients with advanced melanoma ORR: 37% across all doses The median PFS: 7 month The estimated median OS was not reached 79% reported drug-related AEs of any grade 3% reported grade 3 or 4 drug-related AEs
  Robert et al.16 I Intravenous pembrolizumab at 2 mg/kg every or 10 mg/kg every 3 weeks 173 patients with advanced melanoma ORR: 26% at both doses Median PFS: 22 weeks (95% CI 12–36) 2 mg/kg group and 14 weeks (12–24) for the 10 mg/kg group HR 0·84 Most common AEs in any grade were fatigue (33% versus 37%), pruritus (26% versus 19%), and rash (18% versus 18%)
II Intratumoral OncoVex injections followed 3 weeks later by injections every 2 weeks 50 stage IIIC and IV melanoma patients OS > 16 month for all patients. 1 year SR was 58% for all patients and 40% for stage IV M1c patients Grade 1–2 AEs in 85% of patients. Grade 3 AEs infrequent, 21 severe AE unrelated to treatment Virus-based vaccine
I/II Patients were intranodally injected into a tumour-free lymph node 45 stage III and IV melanoma patients PFS: 34·3 month and OS: not reached for stage III patients PFS: 8·1 versus 2·8 month (P: 0·062) in patients with TAA-specific T cells compared with patients without TAA-specific responses, respectively. OS: 24·1 versus 11 month, respectively, for stage IV patients No unexpected or severe events reported Dendritic cell-based vaccine
Breast cancer
III Pertuzumab + trastuzumab + docetaxel. Placebo + trastuzumab + docetaxel As first-line treatment until disease progression or development of toxic events 808 HER2+ breast cancer patients PFS = 18·5 versus 12·4 month Pertuzumab versus placebo group, respectively. HR = 0·62, P < 0·001 AEs incidence at any grade was 5% higher in Pertuzumab group than control. Diarrhoea, rash, mucosal inflammation, febrile neutrophenia and dry skin Monoclonal anti-HER2
  • NCT008299166

  • EMILIA

  • Verma et al.96

III Treatment group received T-DM1 (3·6 mg/kg) intravenously every 21 days Control group self-administered oral lapatinib (1250 mg/day) + oral capecitabine every 12 hr 991 HER2+ breast cancer patients previously treated with trastuzumab plus a taxane PFS: 9·6 versus 6·4 months in T-DM1 and control group, respectively HR: 0·65, P < 0·001 OS: 30·9 versus 25·1 month, respectively HR: 0·68, P < 0·001 ORR: 43·6% and 30·8%, respectively. P < 0·001 Grade ≥ 3 AEs in 57% versus 41% in treatment and control group, respectively Diarrhoea, nausea, vomiting, palmar-plantar erythrodysaesthesia Monoclonal anti-Her2 conjugated to a cytotoxic agent
IIa HER2+ patients treated with T-DM1 (3·6 mg/kg) + pertuzumab (840 mg first time, then 420 mg) once every 3 weeks 64 patients with locally advance or metastatic HER2+ breast cancer Overall ORR: 41% Overall PFS: 6·6% The most frequent grade ≥ 3 AEs were thrombocytopenia, fatigue and liver enzyme elevations Monoclonal anti-Her2 conjugated to a cytotoxic agent
Non-small-cell lung cancer (NSCLC)
II Concurrent arm regimen: ipilimumab (10 mg/kg)/paclitaxel/carboplatin followed by placebo/paclitaxel/carboplatin Phased arm regimen: placebo/paclitaxel/carboplatin followed by ipilimumab (10 mg/kg)/paclitaxel/carboplatin Control regimen: placebo/paclitaxel/carboplatin 204 untreated small-cell lung cancer patients Phased arm, but not concurrent arm, demonstrated an improvement in irPFS compared with chemotherapy (5·7 versus 4·6 months; HR = 0·72; P = 0·05), and OS (12·2 versus 8·3 months) Overall rates of grade 3/4 irAEs: phased arm (15%), concurrent arm (20%) and control (6%) Most common irAE were rash, pruritus and diarrhoea Monoclonal anti-CTLA
II 130 untreated extensive-disease-small-cell lung cancer patients. Phased arm, but not concurrent arm, demonstrastrated an improvement in IRPFS compared with chemotherapy alone (6·4 versus 5·3 months; HR = 0·64; P = 0·03) OS: 12·9 versus 9·9 m Overall rates of grade 3/4 irAEs: phased arm (17%), concurrent arm (21%) and control (9%). Most common irAEs were rash, pruritus and diarrhoea
IIb Patients received up to six cycles of cisplatin–gemcitabine with or without subcutaneous TG4010 each week for 6 weeks 148 advanced NSCLC patients PFS = 43·2% versus 35%, in TG4010 group and chemotherapy alone group, respectively at 6 month ORR = 41·9% versus 28·4%, in TG4010 group and chemotherapy alone group, respectively, at 6 month Abdominal pain, injection-site pain, and fever were more common in patients in the TG4010 plus chemotherapy group than the chemotherapy alone group Antigen associated vaccines
III Patients were randomly assigned to tecemotide and placebo groups. Initial therapy of 8 consecutive weekly subcutaneous injections of tecemotide (806 μg lipopeptide) or placebo every 6 weeks until disease progression 1239 unresectable stage III NSCLC patients No significant difference in OS with the administration of tecemotide after chemoradiotherapy compared with placebo (25·6 versus 22·3 month, respectively) OS = 30·8 versus 20·6 month for tecemotide arm compared with placebo arm, respectively HR = 0·78; P = 0·016 Grade 3–4 AE reported in > 2% frequency with tecemotide were dyspnoea (5% versus 4% in tecemotide versus placebo group), metastases to central nervous system (3% versus 1%), and pneumonia (2% versus 3%) Antigen-associated vaccine
Prostate cancer
II Sipuleucel-T or placebo was intravenously administered every 2 weeks, for a total of 3 infusions 152 metastatic castration-resistant prostate cancer patients Reduction of 22% of risk of death in treatment versus control group. (HR: 0·78, P = 0·03) Median survival 25·8 versus 21·7 month, respectively In treatment group, AEs were chills, fatigue, backpain, hypertension, hypokalaemia and muscular weakness. Only one grade 4 AE Autologous dendritic cell vaccine (mononuclear leucocytes)
II PROSTVAC-VF injections versus control vectors. Priming immunization with rV-PSA-TRICOM with subsequent boosts using rF-PSA-TRICOM, with recombinant GM-CSF 125 castration-resistant metastatic prostate cancer PFS similar in both groups (P = 0·6) OS: 30% versus 17% in treatment and control group, respectively Median survival: 25·1% versus 16·6%, respectively Well tolerated. Most AEs were injection site reactions (swelling, pruritus). General disorders also reported: fatigue, chill and nausea Recombinant vaccinia virus PSA (rV-PSA) vaccine
Renal cell carcinoma (RCC)
I Vaccinations of IMA901, each preceded by administration of GM-CSF as immunomodulator 28 HLA-A*02+ patients with metastatic RCC Patients that responded to multiple tumour-associated peptides were more likely to experience stable disease or partial control (P = 0·019) than subjects that responded to 1 tumour-associated peptides or had no response Well tolerated Multi-peptide vaccine
II Use of single-dose cyclophosphamide (Cy) as additional immunomodulator (+Cy arm) with IMA901 vaccinations
  • 68 HLA-A*02+ patients with metastatic RCC

  • 33 pre-treated (+Cy)

  • 35 not pre-treated (−Cy)

No differences in safety profile in +Cy and −Cy arms PFS = 23·5 versus 14·8 months, respectively (HR: 0·57, P = 0·09) Cy did not alter induction of T cell responses Among immune responders, prolonged survival in +Cy arm. (HR: 0·38, P = 0·04) 2 serious AEs reported: systemic allergic reaction after 12th vaccination (caused by GM-CSF) and grade 3 allergic reaction after 11th vaccination
II Nivolumab (0·3, 2, or 10 mg/kg) was administered intravenously every 3 weeks 168 treated patients with metastatic RCC PFS = 2·7 versus 4 month versus 4·2 month for 0·3-, 2-, and 10-mg/kg groups, respectively ORR = 20%, 22%, and 20% Median OS = 18·2 month, 25·5 month, 24·7 month HR = 0·8, 0·9 in the 2- and 10-mg/kg groups compared with the 0·3-mg/kg group, respectively 73% of patients experienced treatment-related AEs in any grade and 11% experienced a grade 3–4 event Fatigue was the most common treatment-related AE in each group Monoclonal Anti-PD-1

Abbreviations: AE, adverse event; DTIC, dacarbazine; GM-CSF, granulocyte–macrophage colony-stimulating factor; HR, hazard ratio; irAE, immune-related adverse event; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; TAAs, tumour-associated antigens.

Melanoma

Melanoma accounts for the majority of skin-cancer-related deaths. From 2006 to 2010, incidence rates among white people increased by 2·7% per year, and an estimated 76 100 new cases of melanoma were diagnosed in 2014 in the USA.54 Melanoma has been considered as a model tumour for studying anti-tumour immunotherapies because the antigenic profiles are widely characterized for this type of cancer. The first drug treatment against melanoma approved in 1975 by the FDA was dacarbazine (DTIC), an alkyating drug used in chemotherapy that interferes with the proliferation of both cancer and normal cells. In 1998, the use of high-dose IL-2 was approved by the FDA as a treatment for this type of cancer. Nevertheless, although the number of cancer patients treated with IL-2 has rapidly increased since then, there has been a decline in recent years of the use of this therapy because new treatments have been evaluated in recent clinical trials.55 Currently, two new agents for melanoma treatment have been successfully developed: vemurafenib (PLX4032), a drug that inhibits the kinase activity of mutated BRAF gene (present in 40–60% of melanoma patients),56 which has higher affinity for the V600E mutation than the wild-type protein,57 and ipilimumab (MDX-010), a human monoclonal anti-CTLA-4 antibody that blocks T-cell inhibition.

In 2010, Hodi et al.13 showed in a phase III trial (NCT00094653), that ipilimumab improved the overall survival (OS) of 676 patients with unresectable Stage III or Stage IV melanoma who were HLA-A*0201 positive. These patients, who previously received chemotherapy treatment (DTIC and/or temozolomide) with IL-2 doses, were randomly assigned in a 3 : 1 : 1 ratio. The first group was treated with ipilimumab plus a gp100 peptide vaccine. The second group was treated with ipilimumab alone and the third one was treated with gp100 alone. Patients treated with ipilimumab either alone or with gp100 showed improved OS (median OS = 10 and 10·1 months, respectively) compared with gq100 alone (median OS = 6·4 months), indicating that ipilumumab could be administered alone, because it seems to be independent of the gp100 effect. The extended OS of previously treated patients correlated with increased CD8+ T-cell activation and T regulatory (Treg) cell inhibition. The risk of death of these two groups was reduced by 32% (P < 0·001) and 34% (P < 0·003), respectively, compared with the gp100 control group.13 Over 95% of patients in each treatment experiences at least one adverse event (AE) during the trial. The most common immune-related AE (irAE) was tissue inflammation, most frequently in ipilimumab-treated groups. Finally, drug-related deaths occurred in 2·1%, 3·1% and 1·5% in the combined treatment, ipilimumab alone and dp100 alone, respectively. Results of a second phase III trial (NCT00324155) were published by Robert et al.,58 in which 502 patients with previously untreated metastatic melanoma stage IIIC or IV were randomly grouped in a 1 : 1 ratio and treated with ipilimumab plus DTIC or DTIC alone. OS was significantly longer in the group receiving ipilimumab plus DTIC (11·2 months) compared with the control group (9·1 months). The risk of disease progression was reduced by 24% (P < 0·0069) and no drug-related deaths occurred in the ipilimumab group. Nevertheless, this treatment also increases proliferation of autoreactive lymphocytes, increasing the risk of autoimmune disease. Several side effects of the use of ipilimumab have been reported, especially autoimmune-related complications, such as colon perforation, and retinopathy.5964 These side effects should be carefully followed up in future treatments. Notably, this study supported the hypothesis that the combination of chemotherapy with a reduction of Treg cells is a strategy that can provide a more effective anti-tumour immunity.

The success of ipilimumab in melanoma cancer treatment has motivated the use of other antibodies, such as checkpoint receptor inhibitors. As previously mentioned, PD-1 is a key inhibitory receptor expressed by activated T and B cells. It binds to PD-L1 and PD-L2, expressed on antigen-presenting cells and cancer cells. Phase I clinical trials have shown a good safety profile and durable objective tumour regressions in cancer patients to a PD-1 inhibitor, nivolumab (BMS-936558), including melanoma patients65,66 even when combined with ipilimumab,67 reinforcing the idea that combined therapy against CTLA-4 and PD-1 can produce a better anti-tumour response than monotherapy. In a recent phase III clinical trial (NCT00730639), 107 melanoma patients received intravenous nivolumab every 2 weeks for up to 96 weeks; OS rates of 62% at 1 year and 43% at 2 years were reported, and an improved OS (16·8 months). Objective responses were observed in 31% of patients and 7% had disease stabilization lasting at least 6 months. There were no drug-related deaths. Moreover, after 2 years of follow up for some patients after treatment discontinuation, most AEs occurred within the first 6 months of therapy, and cumulative toxicities were not observed with prolonged drug exposure.68 Notably, on 4 September 2014, pembrolizumab (formerly lambrolizumab, MK-3475), was approved by the FDA for treatment of melanoma, being the first anti-PD-1 drug to be approved.69 In 2013, it received the ‘Breakthrough Therapy’ designation from the FDA for treating patients with advanced melanoma due to good results in a phase I clinical trial (NCT01295827).70 In June 2014, at the 50th Annual Meeting of the American Society of Clinical Oncology (ASCO 2014) in Chicago, preliminary results from the largest study performed so far (KEYNOTE-001, NCT01295827) for this drug reported long-term responses. From 411 patients with ipilimumab-refractory and ipilimumab-naive melanoma, 34% met the Response Evaluation Criteria in Solid Tumors (RECIST) objective response criteria, and by October 2013, 88% had sustained their objective response. Even though the median OS has not been reached, 71% of all patients were alive at 1 year.71 These results are even more promising because the most common AEs, in any grade (fatigue, pruritus and rash) affected no more than 1–2% of patients, and only 4% of patients discontinued treatment due to a drug-related side effect. Furthermore, patients presented a unique pattern of responses to pembrolizumab, therefore authors proposed an alternative immune-related response criteria to detect cases that do well clinically, but that under RECIST would not be considered as good clinical responses.72 Results of a randomized expansion cohort of the phase I KEYNOTE-001 study were published. In this study, 173 patients with unresectable melanoma who were previously treated with at least two doses of ipilimumab and had confirmed disease progression were recruited.16 Also, patients with BRAF mutations that were treated with approved BRAF and/or MEK inhibitors were included. Patients received either pembrolizumab 2 or 10 mg/kg and were followed for a median time of 8 months. Objective response rate was 26% at both doses (difference 0%, P = 0·96). Median PFS was 22 weeks for the pembrolizumab 2 mg/kg group and 14 weeks for the pembrolizumab 10 mg/kg group [Hazard Ratio (HR) 0·84, 95% CI 0·57–1·23]. Grade 3 or 4 drug-related AEs occurred in only 12% patients and grade 3 or 4 irAE occurred in only three patients. These results reflect a good option for the treatment for ipilimumab-refractory melanoma patients, which do not have other treatment options so far. Also, a phase II study is currently ongoing, the KEYNOTE-002 study (NCT01704287), in which the treatment is being tested on 540 advanced melanoma patients.16

Allovectin-7, a gene-therapy-based immunotherapy, was designed to boost both adaptive and innate immune responses. It is a bicistronic plasmid/lipid composite that encodes the human leucocyte antigen-B7 heavy-chain gene and the β2-microglobulin gene. It is locally injected and promotes the allogeneic expression of MHC-I antigen, which is usually lost in melanoma cancer cells.73 In 2010, a phase II clinical trial (NCT00044356) reported that 11·8% of selected stage III/IV metastatic melanoma patients achieved an objective response with median duration of 13·8 months and median OS of 18·8 months.74 Nevertheless, two phase III clinical trials (NCT00395070, NCT00003647) failed to reach the primary and secondary efficiency end-points. As well as Allovectin-7, OncoVEXGM-CSF (Talimogene laherparepvec, T-VEC), is an intralesional therapy that consists of recombinant herpes simplex virus type 1 encoding for GM-CSF. It selectively enters cancer cells75 and expresses GM-CSF, inducing cell lysis. Cell debris stimulates the maturation, proliferation and differentiation of dendritic cells, so stimulating a tumour-specific immune response. Results of a phase II clinical trial (NCT00289016) were published in 2009, in which 50 patients, 10 with stage IIIC disease and 40 with stage IV disease (74% of them had previously received dacarbazine/temozolomide or IL-2 pre-treatment) were locally injected and a 26% ORR was obtained. Eight had complete response and five had a partial response; OS was 58% at 1 year and 52% at 24 months.76 Regressions of distant lesions were documented. Also, post-treatment phenotypic evaluation of tissue samples revealed that numbers of Treg cells, CD8+ T cells and myeloid-derived suppressor cells were higher in the tumour environment than in peripheral blood, as is usually seen for melanoma patients. The accumulation of these suppressor cells was decreased upon treatment with OncoVEXGM-CSF, and also induced local and systemic MART-1-specific CD8+ effector cells.77 Due the good results obtained, there is a phase III clinical trial ongoing78 (NCT00769704) to evaluate safety and risks of this treatment.

Messenger RNA-transfected DCs have been shown to be the most effective loading strategy to induce immune responses because transfected mRNA can produce multiple and possibly more immunogenic antigenic epitopes than an antigen-loading strategy.79 It is also independent of the patient's HLA haplotype, as HLA restriction is the main limitation for use of peptides as antigens for DC priming. Besides, mRNA insertion into DCs is considered to be safe and clinically applicable because it has a short half-life and does not insert into the host genome,80 and can be used for expressing co-stimulatory molecules into DCs such as toll-like receptors.81 Since the first immunotherapy pilot clinical trial performed in 1998 in melanoma patients using DC-based vaccines,82 several clinical trials are now ongoing. Aarntzen et al.83 presented a phase I and phase II clinical trial (NCT00243529) in 45 stage III and stage IV melanoma patients who were vaccinated with autologous mature monocyte-derived DCs electroporated with mRNA coding for the TAAs gp100 and tyrosinase protein, and pulsed with keyhole limpet haemocyanin protein. There was an enhancement of tumour-specific CD8+ and CD4+ T-cell responses and enhanced production of interferon-γ in stage II patients. In stage III patients, the median PFS was 34·3 months and in Stage IV patients, those that presented TAA-specific T cells tended toward an improved PFS (8·1 months) and OS (24·1 months) compared with patients without TAA-specific responses (PFS = 2·8 months; P = 0·062 and OS = 11 months; P = 0·101). These results presented DC-based vaccination as a promising adjuvant treatment of stage III melanoma patients. The same year, Oshita et al. published the results for a phase II clinical trial (number not reported) in Japanese patients, in which monocyte-enriched fractions were pulsed with a cocktail of five melanoma-specific synthetic peptides restricted to HLA-A2 or A24 (MART-1, gp100, tyrosinase, MAGE-A2, and MAGE-A3; and gp100, tyrosinase, MAGE-A1, MAGE-A2 and MAGE-A3, respectively) and keyhole limpet haemocyanin. Overall survival in the vaccinated group was higher (13·6 months) when compared with non-vaccinated patients (7·3 months).84

Breast cancer

HER2 is a transmembrane tyrosine kinase receptor with no direct ligand identified. It is associated with activation of the phosphoinositide 3-kinase and AKT pathways, regulating a complex signal transduction cascade that influences cell cycle progression, cell survival, cell proliferation and cell motility.85,86 Over-expression of the HER2 oncogene, shown to lead malignant transformation of cells, account for 20–30% of all breast cancer cases.87 In 1998, trastuzumab (Herceptin), a monoclonal antibody that binds to domain IV of the extracellular domain of the HER2 receptor, was approved by the FDA for treatment of HER2-positive breast cancer patients and nowadays it is the standard treatment for these patients. This was the first monoclonal antibody approved by the FDA for the treatment of solid tumours. Treatment with trastuzumab implies the direct effect of blocking the HER2 constitutive function, and also prevents cleavage of the HER2 extracellular domain,88 which is an extra mechanism for HER2 signalling activation.85 Also, indirect effects on inflammatory response take place; leading to antibody-dependent cell-mediated cytotoxicity, and so the induction of an adaptive immune response.8991 A 6-year follow-up phase III clinical trial (NCCTG N983) demonstrated that concurrent trastuzumab treatment together with chemotherapy has improved disease-free survival and a better outcome.92 Also, results suggested that the mechanism of action of trastuzumab also intensifies chemotherapy-induced cytotoxicity.93 New therapeutic monoclonal antibodies have been developed to increase or complement the effect of trastuzumab, such as Pertuzumab, a humanized monoclonal antibody that binds domain II of HER2. This domain is known to dimerize with other HER receptors, especially with HER3, potentiating together the HER2 oncoactivity in tumours over-expressing HER2.85 Baselga et al.94 published the results of the first phase III clinical trial (CLEOPATRA, NCT00567190) in which trastuzumab and pertuzumab plus docetaxel combined therapy was evaluated in 808 patients with HER2+ metastatic breast cancer. They were randomly grouped in a 1 : 1 ratio; one group received trastuzumab plus docetaxel and the other received trastuzumab plus pertuzumab plus docetaxel. The median PFS was improved by 6·1 months, 12·4 months in the control group, as compared with 18·5 months in the pertuzumab group (P < 0·001), and a reduced risk of progression or death (HR = 0·62) was reported. Importantly, even though increased rates of cardiac dysfunction with trastuzumab have been reported in earlier clinical trials, the combination of pertuzumab plus trastuzumab plus docetaxel did not increase the incidence of cardiac AEs compared with the control group. The benefits of combined targeting of HER2 reported in CLEOPATRA were later confirmed after a one additional year follow up.95 Despite the success of these therapies for HER2-positive metastatic breast cancer treatment, still many patients eventually progress and or present severe toxic side effects and an effort to develop more efficient and less toxic therapies was necessary. EMILIA, a phase III clinical trial (NCT00829166) evaluated the efficacy and safety of trastuzumab emtansine (T-DM1), a monoclonal anti-HER2 antibody linked to a microtubule-inhibitory agent, mertansine (DM1), in patients with HER2-positive advanced breast cancer who have previously received trastuzumab and one of the taxane chemotherapies.96 This methodology allowed delivery of chemotherapy directly to HER2-over-expressing cells. Nine hundred and ninety-one patients were randomly assigned in a 1 : 1 ratio to T-DM1 (n = 495) or lapatinib plus capecitabine (n = 496). Treatment with T-DM1 improved PFS, with a median survival of 9·6 months compared with 6·4 months with lapatinib plus capecitabine, and improved OS of 30·9 versus 25·1 months, respectively. These results allowed T-DM1 to be approved by the FDA the same year for patients with HER2-positive, metastatic breast cancer who have previously received trastuzumab or one of the taxane chemotherapies (paclitaxel or docetaxel). Second end-point studies have supporter the high efficacy and tolerability of T-DM1 for the treatment of women with HER2-positive breast cancer.97 Recently, a phase IIa clinical trial (NCT00875979) has reported the safety and efficacy of the combined antibody therapy using T-DM1 plus pertuzumab in HER2-positive locally advanced breast cancer or metastatic breast cancer patients.98 They reported an objective response rate of 41% (26 of 64 patients) and a median PFS of 6·6 months, along with an acceptable safety profile. The phase III clinical trial (MARIANNE, NCT01120184) is now evaluating safety and efficacy of T-DM1 plus pertuzumab versus T-DM1 plus placebo versus trastuzumab plus a taxane in patients with HER2-positive, progressive, or recurrent locally advanced or chemotherapy-naive metastatic breast cancer.

In addition to the immunotherapy approaches mentioned above, other groups are focused on the development of vaccines that may improve the immune response against a specific target, providing a less cytotoxic effect on non-malignant cells. HER2 peptides, such as E7 (amino acids 369–377 = KIFGSLAFL, also known as NeuVax) and GP2 (amino acids 654–662 = IISAVVGIL) have shown high immunogenicity and its use for anti-cancer vaccines has been proposed and tested. In a phase II study in 195 patients, after 24 months of E7 vaccine treatment, the treated group (E7 plus GM-CSF) showed a 57% reduction in relative risk of recurrence compared with the control group, suggesting that E7 vaccine treatment is a safe and effective option for stimulation of HER2-specific immunity.99 Given that it has been previously described, using tumour cell line-based experiments, that trastuzumab and GP2 might act synergistically to stimulate cytotoxic T cells ex vivo,100 the authors suggest that GP2 vaccination during trastuzumab treatment might be an efficient complementary anti-cancer treatment. In addition, in 2010, Carmichael et al. presented the first phase I trial in which GP2 vaccine was tested in 18 breast cancer patients to document safety, immunological responses and epitope spreading. The drug was shown to be safe and well tolerated.101 Currently, there is a phase II clinical trial (NCT00524277) evaluating the efficacy of GP2 compared with GM-CSF in treating patients with breast cancer.

Cryoablation is a procedure that has already been successfully tested in breast cancer patients.102,103 It involves the insertion of a cryoprobe into the tumour monitored by a magnetic resonance imaging system, and the tissues are then frozen in vivo to kill cancer cells. Afterwards, broken cells will be recognized by the patient's immune system.104 In addition, patients are treated with ipilimumab to keep T cells active and to induce memory T cells to prevent cancer returning in the future. It is a promissory approximation because it has been demonstrated that mice with breast cancer show increased production of tumour-specific T cells, eradication of micrometastasis and a better outcome after cryoablation depending on the freezing methods used.104,105 There is an ongoing pilot study (NCT01502592) for a small group of pre-operative early stage/resectable breast cancer patients, in which the safety of a combined strategy based on magnetic resonance imaging-guided cryoablation and immunotherapy will be evaluated.

Non-small-cell lung cancer

Lung cancer is one of the leading causes of death by cancer in men and women worldwide. NSCLC comprises approximately 84% of all lung cancer cases.106 Although NSCLC was thought to be poorly immunogenic, recent clinical trials testing checkpoint inhibitors and cancer vaccines have shown promising results, and these approaches are now in late-phase development. Given the positive results obtained for ipilimumab in melanoma,107 this drug was tested in a phase II clinical trial (NCT00527735). In 204 previously untreated NSCLC patients it was used together with carboplatin and placlitaxel.108 The treatment improved ipilimumab efficacy in tumours with squamous histology in patients under a phased ipilimumab regimen when compared with a concurrent ipilimumab regimen. In the phased arm both PFS and OS improved (PFS = 5·7 versus 4·6 months; HR, 0·72; P = 0·05, and OS = 12·2 versus 8·3 months, as compared with paclitaxel and carboplatin alone). In parallel, Reck et al.109 presented the results for 130 extensive-disease (ED-) NSCLC from the same clinical trial. There was an improvement in PFS in patients treated with the phased ipilimumab regimen, but not in those given concurrent ipilimumab, compared with chemotherapy alone (6·4 versus 5·3 months; HR = 0·64; P = 0·03) and an improved OS (12·9 versus 9·9 months). In both studies, irAEs involved skin (rash and pruritus) and gastrointestinal tract (diarrhoea), occurring more frequently in patients treated with ipilimumab. After these results, two phase III clinical trials are in progress to test the phased regimen with ipilimumab in contrast to standard chemotherapy in NSCLC and ED-NSCLC patients (NCT01285609 and NCT01450761, respectively).

Regarding anti-PD-1 treatment, a phase I clinical trial has demonstrated promissory results. Nivolumab was tested for activity in 122 NSCLC using a dose escalation method of 1·0, 3·0, or 10 mg/kg every 2 weeks in an 8-week cycle. ORR was observed in 33% of patients with squamous histology and 12% of patients with non-squamous histology.37 Two phase III clinical trials comparing nivolumab with docetaxel in patients with squamous and non-squamous NSCLC (NCT01642004 and NCT01673867; respectively) are currently active.

MUC1 is a highly glycosylated transmembrane protein that is expressed on the apical membrane of normal cells110 and aberrantly expressed and glycoslated in tumour cells, exposing tumour-associated epitopes.111 TG4010 is an antigen-based vaccine composed of the genetically modified vaccinia virus Ankara containing the MUC1 sequence and IL-2, which acts as adjuvant. Results from a phase IIB clinical trial (NCT00415818) were published by Quoix et al.112 One hundred and forty-eight patients with advanced NSCLC, positive for MUC1 expression by immunohistochemistry, were separated in two groups. Group 1 was treated with chemotherapy in combination with TG4010 and group 2 was treated with chemotherapy alone. After 6 months of treatment, PFS and ORR were higher in the TG4010 plus chemotherapy group (PFS = 43·2%; ORR = 41·9%) compared with the chemotherapy-alone group (PFS = 35%, ORR = 28·4%). There were no significant differences of median OS between both groups; however, median overall survival for patients who presented an objective response was higher in the TG4010 plus chemotherapy group compared with the chemotherapy alone group (23·3 versus 12·5 months). In general, the most common AEs in both groups were anaemia, neutropenia and thrombocytopenia. Abdominal pain, injection-site pain and fever were more common in patients in the TG4010 plus chemotherapy group than the chemotherapy alone group. These results suggest that TG4010 is able to improve first-line treatment in advanced NSCLC. Tecemotide (L-BLP25) is a MUC1-derived 25-amino-acid BLP25 lipopeptide vaccine that was tested in a phase III trial (START, NCT00409188). In this study, 829 and 410 patients were randomly assigned to tecemotide and placebo group, respectively.113 Although the drug failed to meet the primary end-points regarding OS, where 25·6 months were reported in the tecemotide arm, compared with 22·3 months in the placebo group (HR = 0·88; P = 0·12), interesting results were obtained in a subset of patients receiving initial concurrent chemoradiotherapy. OS was higher for patients that received tecemotide compared with patients of the placebo arm (OS = 30·8 versus 20·6 months; respectively; HR = 0·78; P = 0·016). No statistical differences were observed between tecemotide and placebo for any AE. The clinical benefit obtained in the subgroup of patients that received concurrent chemoradiotherapy was not observed previously in other approaches against NSCLC. The START2 clinical trial will assess the efficacy, safety and tolerability of tecemotide in patients with unresectable advanced NSCLC who have showed a response or stable disease after platinum-based concurrent chemoradiotherapy.

Prostate and renal cell carcinoma

Prostate-specific antigen (PSA) is expressed by prostate cells, and naturally specific T helper type 1 responses to this antigen have been found in patients with metastatic prostate cancer.114 However, the prevalence and magnitude of detectable responses to PSA are low and clinically insignificant.114 Under the concept that the low response to the antigen can be positively modulated, vaccines have been designed to improve the immune recognition of this weak immunogenic antigen and induce a specific T-cell response. Among the cell-based vaccines tested so far, only one has been approved by the FDA for the treatment of asymptomatic and minimally symptomatic metastatic castration-resistant prostate cancer, sipuleucel-T, a DC-based vaccine that demonstrated successful efficacy in phase III clinical trials.14,115,116 This immunostimulant treatment was catalogued as ‘category 1’ or highly recommended vaccine for the treatment of hormone-refractory prostate cancer. The largest Phase III clinical trial performed is IMPACT15 (NCT00065442), where 512 patients were assigned in a 2 : 1 ratio. One group (n = 341) received sipuleucel-T and the other (n = 171) received placebo, both administered intravenously every 2 weeks. There was a relative reduction of 22% in the risk of death in the sipuleucel-T group as compared with the placebo group (HR = 0·78). This reduction represented a 4·1-month improvement in median survival (25·8 versus 21·7 months, respectively). There is an ongoing Phase I clinical trial (NCT01832870) designed to quantify the immune response and determine the tolerability of sipuleucel-T when given in combination with ipilimumab for patients with advanced prostate cancer. Also, a phase III clinical (NCT01322490) trial is currently ongoing for a poxviral-based PSA vaccine, PROSTVAC-VF (PSA-TRICOM). Previous results for a phase II clinical trial were published in 2010 (NCT00078585),117 in which 82 patients received PROSTVAC and 40 received control vectors. After 3 years of treatment, OS was higher for PROSTVAC-treated patients (25·1 versus 16·6 months in control group), although the study failed to find an association between PROSTVAC-treated patients and PFS.

Renal-cell carcinoma is the most frequent type of kidney cancer, accounting for 90% of adult cases although the incidence varies with geographic location worldwide.118 Walter et al., have developed a tumour-associated peptide-based vaccine named IMA901 through a discovery platform called XPRESIDENT that allowed the detection of nine naturally expressed tumour-specific antigens from primary renal tumour tissues. The vaccine was tested in HLA-A*02+ patients with advanced RCC in a phase II clinical trial (NCT00523159) in which T-cell responses to IMA901 and clinical benefit association were assessed.31 The treatment was safe and well tolerated, although there were two cases of serious AEs associated with allergic reactions. There was a prolonged survival in patients treated with IMA901 and pre-treated with cyclophosphamide (OS = 23·5 months) versus non-pre-treated (OS = 14·8 months, HR = 0·57; P = 0·09) and an association was established between immune response and OS because there was a prolonged survival among immune responders pre-treated with cyclophosphamide compared with subjects without this pre-treatment (HR = 0·38; P = 0·040). Finally, MDSC4 and MDSC5, phenotypes for myeloid-derived suppressor cells, were negatively associated with OS, and T-cell receptor-ζ expression tended to associate positively, suggesting them as cellular biomarkers. APOA1 and CCL17 were suggested as serum biomarkers for vaccine-induced immunity and OS. Now, a phase III clinical trial (IMPRINT, NCT01265901) is ongoing to assess whether IMA901 can prolong OS in patients with metastatic and/or locally advanced RCC when given along with sunitinib, a tyrosine kinase inhibitor. This multiantigen-based vaccine is a promissory approach for RCC treatment because other vaccines, such as Reniale, an autologous RCC tumour cell lysate; Vitespen an autologous tumour-derived heat-shock protein Gp96 preparation; and Trovax, a single-tumour antigen vaccine, have not been approved for clinical use because phase III clinical trials have not reached the expected results.119

Several studies have shown that PD-L1 expression in RCC tumours is associated with rapid progression and poor outcomes.120122 On the other hand, PD-1 is expressed in TILs from clear-cell RCC and has been described as an indicator of poor prognosis, poor survival and as a predictor of latent distant metastasis of clear-cell RCC.123,124 Results of a recent phase II trial (NCT01354431) in which nivolumab was tested for activity and safety in previously treated patients with metastatic RCC have been published.125 One hundred and sixty-eight patients were grouped in a 1 : 1 : 1 ratio and received nivolumab (0·3, 2, or 10 mg/kg) administered intravenously every 3 weeks. Median PFS was 2·7 months (80%), 4·0 months (80%), and 4·2 months (80%) and ORR was 20% (n = 12), 22% (n = 12), and 20% (n = 11) in the 0·3-, 2-, and 10-mg/kg groups, respectively. The median OS was 18·2, 25·5 and 24·7 months in the 0·3-, 2-, and 10-mg/kg groups, respectively, within a minimum follow up of 24 months. Seventy-three per cent of patients experienced treatment-related AEs in any grade and 11% experienced a grade 3–4 event. Forty per cent of patients had received two or three previous anti-angiogenic drugs and one-third had received everolimus previously. In this regard, two phase III clinical trials are currently evaluating metastatic RCC patients. One of them is focused on comparing nivolumab with everolimus in patients pre-treated with anti-angiogenic therapy and the other is assesing the combination of nivolumab and ipilimumab (NCT01668784 and NCT02231749, respectively).

  1. Persisting challenges for cancer immunotherapy due patient heterogeneity

It is evident that the therapies discussed above still fail to improve OS for a high percentage of patients, bringing a small benefit or even only adverse events. The question as to why immunotherapeutic approaches work in some patients and fail in others remains unanswered. Every tumour develops distinct mechanisms to evade the immune system, which could confer special resistance to certain immunotherapy. Efforts are being made to identify the subsets of patients in which immunotherapy could work more efficiently. The main strategies used so far are the detection of prognostic and predictive biomarkers through gene expression profiling,126 as well as immune peptidome analyses of HLA-bound peptides and epitope prediction by informatics approaches for TAA identification. Notably, a recent study has suggested why ipilimumab works for some patients and not for others by analysing mutation-derived antigen identification through whole exome sequencing of DNA from tumour cells.127 The results showed that tumours of ipilimumab responders have a higher mutational burden than those from patients that did not respond to the therapy. A higher mutational rate gives rise to a higher number of neoantigens that can activate T cells after CTLA-4 blockade. However, not all tumours with a high mutation burden responded to the therapy. These studies were extended by using bioinformatic tools to identify among the responders group a subset of antigens that could elicit an augmented anti-tumour immune response in these patients.127 Interestingly, whole exome-sequencing and bioinformatic approaches were used to determine neoantigens in mouse models, leading to the identification of tumour-specific neoantigens that, when used to immunize mice, induce T-cell responses that rejected established tumours.128 Immunization with tumour neoantigens together with targeting PD-1 or CTLA-4 can induce T-cell activation and rejection of tumours expressing those neoantigens.129 These results highlight the importance of using new methodologies, such as whole exome sequencing and bioinformatic tools to identify neoepitopes for the development of personalized immunotherapy that could improve clinical benefit in non-responder patients.

It should be considered when testing new therapies that not all patients manifest a specific clinical response at the same time and with the same magnitude. The most used response evaluation criteria, RECIST or World Health Organization, which determine patient's response, sometimes fail to define whether a new treatment is really efficient because they do not consider the initial immune response effect on the tumour. For example, an increase in tumour burden followed by a reduction or the appearance of new tumour lesions as a product of the reclusion of immune cells to the tumours would be considered as progressive disease.130 To consider these alternative immune-response-related scenarios, a new criterion has been proposed – the immune-related response criteria.130 These criteria allow the recognition of optimal responses that otherwise would not be recognized by the two classical response evaluation criteria. In spite of this, many recent works still use RECIST for patient's response evaluation.131 Moreover, the identification of biomarkers for treatment response and others that could predict a patient's response to a given treatment is an issue that must also be considered to determine the efficiency of treatment and to avoid cytotoxic effects and low response ratios, respectively.

Concluding remarks

Even though our knowledge of the complexity of the immune cell interactions that underlie the complete immune response against tumour antigens is still far from complete, new therapies have successfully demonstrated their ability to down-modulate tumour progression through intervening or improving different steps of the immune response process.

Tumour microenvironment must not be ignored. Different cell types cooperatively establish the tumour immune profile;40 this is why immunotherapy must not only look for tumour properties to be targeted but also modulate all features as far as possible. Combined treatments of chemotherapy plus immunotherapy have so far shown a better response and OS in the treatment of many cancer types. However, there is still much to do to improve the management of immune responses because there are plenty of promising therapies focused on the exclusive attack of cancer cells, avoiding the systemic toxicity produced by general drugs. The use of combined treatment of chemotherapy plus immunotherapy has been extensively discussed and studied.58,94 The principal goal of these combined approaches is not only to alter the internal signalling pathways of cancer cells, but also to target extrinsic cells and mediators of cancer cell proliferation. Combined therapy has shown excellent results improving overall survival and tumour response in clinical trials, becoming the best approach for cancer treatment.

Disclosures

Authors declare no competitive interests.

Glossary

Abbreviations:

AE

adverse event

CTLA-4

T-lymphocyte-associated antigen 4

DC

dendritic cell

DTIC

dacarbazine

FDA

Food and Drug Administration

GM-CSF

granulocyte–macrophage colony-stimulating factor

HER2

human epidermal growth factor receptor 2

HR

hazard ratio

IL-2

interleukin-2

irAE

immune-related AE

LAG-3

lymphocyte activation gene 3

NSCLC

non-small cell lung cancer

NY-ESO-1

cancer-testis antigen

ORR

objective response rate

OS

overall survival

PD-1

programmed cell death 1

PD-L1

programmed death-ligand 1

PFS

progression-free survival

PSA

prostate-specific antigen

RCC

renal-cell cancer

RECIST

Response Evaluation Criteria in Solid Tumours

siRNA

small interfering RNA

TAAs

tumour-associated antigens

T-DM1

trastuzumab emtansine

TILs

tumour-infiltrating lymphocytes

Treg

T regulatory cells

References

  1. DeVita VT, Rosenberg SA. Two hundred years of cancer research. N Engl J Med. 2012;366:2207–14. doi: 10.1056/NEJMra1204479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309–22. doi: 10.1016/j.ccr.2012.02.022. [DOI] [PubMed] [Google Scholar]
  3. Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol. 2006;1:119–50. doi: 10.1146/annurev.pathol.1.110304.100224. [DOI] [PubMed] [Google Scholar]
  4. Evans A, Costello E. The role of inflammatory cells in fostering pancreatic cancer cell growth and invasion. Front Physiol. 2012;3:270. doi: 10.3389/fphys.2012.00270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Deschoolmeester V, Baay M, Van Marck E, Weyler J, Vermeulen P, Lardon F, Vermorken JB. Tumor infiltrating lymphocytes: an intriguing player in the survival of colorectal cancer patients. BMC Immunol. 2010;11:19. doi: 10.1186/1471-2172-11-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. DeNardo DG, Brennan DJ, Rexhepaj E, et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011;1:54–67. doi: 10.1158/2159-8274.CD-10-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Ruffell B, Au A, Rugo HS, Esserman LJ, Hwang ES, Coussens LM. Leukocyte composition of human breast cancer. Proc Natl Acad Sci USA. 2012;109:2796–801. doi: 10.1073/pnas.1104303108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini P-L. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother. 2011;60:319–26. doi: 10.1007/s00262-010-0968-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Mohammed ZM, Going JJ, Edwards J, Elsberger B, Doughty JC, McMillan DC. The relationship between components of tumour inflammatory cell infiltrate and clinicopathological factors and survival in patients with primary operable invasive ductal breast cancer. Br J Cancer. 2012;107:864–73. doi: 10.1038/bjc.2012.347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ascierto PA, Capone M, Urba WJ, et al. The additional facet of immunoscore: immunoprofiling as a possible predictive tool for cancer treatment. J Transl Med. 2013;11:54. doi: 10.1186/1479-5876-11-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. NCI dictionary of cancer terms – National Cancer Institute [Internet] [WWW document]. URL http://www.cancer.gov/dictionary?cdrid=45729 [accessed on 7 September 2014]
  12. Rosenberg SA. IL-2: the first effective immunotherapy for human cancer. J Immunol. 2014;192:5451–8. doi: 10.4049/jimmunol.1490019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cheever MA, Higano CS. PROVENGE (Sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine. Clin Cancer Res. 2011;17:3520–6. doi: 10.1158/1078-0432.CCR-10-3126. [DOI] [PubMed] [Google Scholar]
  15. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  16. Robert C, Ribas A, Wolchok JD, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–17. doi: 10.1016/S0140-6736(14)60958-2. [DOI] [PubMed] [Google Scholar]
  17. Monjazeb AM, Hsiao H-H, Sckisel GD, Murphy WJ. The role of antigen-specific and non-specific immunotherapy in the treatment of cancer. J Immunotoxicol. 2012;9:248–58. doi: 10.3109/1547691X.2012.685527. [DOI] [PubMed] [Google Scholar]
  18. Iezzi M, Quaglino E, Amici A, Lollini P-L, Forni G, Cavallo F. DNA vaccination against oncoantigens: a promise. Oncoimmunology. 2012;1:316–25. doi: 10.4161/onci.19127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Mann JR, Lakin GE, Leonard JC, et al. Clinical applications of serum carcinoembryonic antigen and alpha-fetoprotein levels in children with solid tumours. Arch Dis Child. 1978;53:366–74. doi: 10.1136/adc.53.5.366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Qian X, Li C, Pang B, Xue M, Wang J, Zhou J. Spondin-2 (SPON2), a more prostate-cancer-specific diagnostic biomarker. PLoS ONE. 2012;7:e37225. doi: 10.1371/journal.pone.0037225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Ryu YJ, Kim CH, Kim HJ, et al. Clinical significance of serial serum carcinoembryonic antigen values for treating rectal cancer with preoperative chemoradiotherapy. J Korean Soc Coloproctol. 2012;28:205–12. doi: 10.3393/jksc.2012.28.4.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Yang L, Chen X, Li Y, Yang J, Tang L. Declines in serum CYFRA21-1 and carcinoembryonic antigen as predictors of chemotherapy response and survival in patients with advanced non-small cell lung cancer. Exp Ther Med. 2012;4:243–8. doi: 10.3892/etm.2012.570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Aggarwal C, Meropol NJ, Punt CJ, et al. Relationship among circulating tumor cells, CEA and overall survival in patients with metastatic colorectal cancer. Ann Oncol. 2013;24:420–8. doi: 10.1093/annonc/mds336. [DOI] [PubMed] [Google Scholar]
  24. Yang C, Ruffner MA, Kim S-H, Robbins PD. Plasma-derived MHC class II+ exosomes from tumor-bearing mice suppress tumor antigen-specific immune responses. Eur J Immunol. 2012;42:1778–84. doi: 10.1002/eji.201141978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Ciravolo V, Huber V, Ghedini GC, et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J Cell Physiol. 2012;227:658–67. doi: 10.1002/jcp.22773. [DOI] [PubMed] [Google Scholar]
  26. Aung T, Chapuy B, Vogel D, et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc Natl Acad Sci U S A. 2011;108:15336–41. doi: 10.1073/pnas.1102855108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Marleau AM, Chen C-S, Joyce JA, Tullis RH. Exosome removal as a therapeutic adjuvant in cancer. J Transl Med. 2012;10:134. doi: 10.1186/1479-5876-10-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Gnjatic S, Nishikawa H, Jungbluth AA, et al. NY-ESO-1: review of an immunogenic tumor antigen. Adv Cancer Res. 2006;95:1–30. doi: 10.1016/S0065-230X(06)95001-5. [DOI] [PubMed] [Google Scholar]
  29. Noguchi T, Kato T, Wang L, et al. Intracellular tumor-associated antigens represent effective targets for passive immunotherapy. Cancer Res. 2012;72:1672–82. doi: 10.1158/0008-5472.CAN-11-3072. [DOI] [PubMed] [Google Scholar]
  30. Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–37. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18:1254–61. doi: 10.1038/nm.2883. [DOI] [PubMed] [Google Scholar]
  32. Bossi G, Buisson S, Oates J, Jakobsen BK, Hassan NJ. ImmTAC-redirected tumour cell killing induces and potentiates antigen cross-presentation by dendritic cells. Cancer Immunol Immunother. 2014;63:437–48. doi: 10.1007/s00262-014-1525-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Singh-Jasuja H, Emmerich NPN, Rammensee H-G. The Tübingen approach: identification, selection, and validation of tumor-associated HLA peptides for cancer therapy. Cancer Immunol Immunother. 2004;53:187–95. doi: 10.1007/s00262-003-0480-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Hassan NJ, Adams K, Bossi G, et al. Abstract 3525: IMCgp100: a novel bi-specific biologic for the treatment of malignant melanoma. Cancer Res. 2012;72(8 Suppl):3525. [Google Scholar]
  35. Salem ML, Rubinstein MP. Cole DJ. Tumours : Immunotherapy. 2007 ;. doi: 10.1002/9780470015902.a0001432.pub2. [Google Scholar]
  36. Bensinger WI. Allogeneic transplantation: peripheral blood vs. bone marrow. Curr Opin Oncol. 2012;24:191–6. doi: 10.1097/CCO.0b013e32834f5c27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010;236:219–42. doi: 10.1111/j.1600-065X.2010.00923.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21:137–48. doi: 10.1016/j.immuni.2004.07.017. [DOI] [PubMed] [Google Scholar]
  41. Powles T, Eder JP, Fine GD, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:558–62. doi: 10.1038/nature13904. [DOI] [PubMed] [Google Scholar]
  42. Herbst RS, Soria J-C, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7. doi: 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Lu J, Lee-Gabel L, Nadeau MC, Ferencz TM, Soefje SA. Clinical evaluation of compounds targeting PD-1/PD-L1 pathway for cancer immunotherapy. J Oncol Pharm Pract. 2014 doi: 10.1177/1078155214538087. ; doi: 10.1177/1078155214538087. [DOI] [PubMed] [Google Scholar]
  44. Fourcade J, Sun Z, Benallaoua M, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207:2175–86. doi: 10.1084/jem.20100637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A. 2010;107:7875–80. doi: 10.1073/pnas.1003345107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Baitsch L, Baumgaertner P, Devêvre E, et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J Clin Invest. 2011;121:2350–60. doi: 10.1172/JCI46102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207:2187–94. doi: 10.1084/jem.20100643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Huard B, Tournier M, Hercend T, Triebel F, Faure F. Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes. Eur J Immunol. 1994;24:3216–21. doi: 10.1002/eji.1830241246. [DOI] [PubMed] [Google Scholar]
  49. Grosso JF, Kelleher CC, Harris TJ, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest. 2007;117:3383–92. doi: 10.1172/JCI31184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Woo S-R, Turnis ME, Goldberg MV, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72:917–27. doi: 10.1158/0008-5472.CAN-11-1620. NIH Public Access. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Shiao SL, Ganesan AP, Rugo HS, Coussens LM. Immune microenvironments in solid tumors: new targets for therapy. Genes Dev. 2011;25:2559–72. doi: 10.1101/gad.169029.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hobo W, Novobrantseva TI, Fredrix H, et al. Improving dendritic cell vaccine immunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles combined with antigen mRNA electroporation. Cancer Immunol Immunother. 2013;62:285–97. doi: 10.1007/s00262-012-1334-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Yu Z, Qian J, Wu J, Gao J, Zhang M. Allogeneic mRNA-based electrotransfection of autologous dendritic cells and specific antitumor effects against osteosarcoma in rats. Med Oncol. 2012;29:3440–8. doi: 10.1007/s12032-012-0312-y. [DOI] [PubMed] [Google Scholar]
  54. Cancer treatment & survivorship facts & figures: 2014–2015 acspc-042801.pdf [Internet]. [WWW document]. URL http://www.cancer.org/acs/groups/content/@research/documents/document/acspc-042801.pdf [accessed on 2 January 2015]
  55. Dillman RO, Barth NM, VanderMolen LA, Mahdavi K, McClure SE. Should high-dose interleukin-2 still be the preferred treatment for patients with metastatic melanoma? Cancer Biother Radiopharm. 2012;27:337–43. doi: 10.1089/cbr.2012.1220. [DOI] [PubMed] [Google Scholar]
  56. Sala E, Mologni L, Truffa S, Gaetano C, Bollag GE, Gambacorti-Passerini C. BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells. Mol Cancer Res. 2008;6:751–9. doi: 10.1158/1541-7786.MCR-07-2001. [DOI] [PubMed] [Google Scholar]
  57. Bollag G, Hirth P, Tsai J, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 2010;467:596–9. doi: 10.1038/nature09454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
  59. Audemard A, de Raucourt S, Miocque S, et al. Melanoma-associated retinopathy treated with ipilimumab therapy. Dermatology. 2013;227:146–9. doi: 10.1159/000353408. [DOI] [PubMed] [Google Scholar]
  60. Dilling P, Walczak J, Pikiel P, Kruszewski WJ. Multiple colon perforation as a fatal complication during treatment of metastatic melanoma with ipilimumab – case report. Pol Przegl Chir. 2014;86:94–6. doi: 10.2478/pjs-2014-0017. [DOI] [PubMed] [Google Scholar]
  61. Du Rusquec P, Saint-Jean M, Brocard A, Peuvrel L, Khammari A, Quéreux G, et al. Ipilimumab-induced autoimmune pancytopenia in a case of metastatic melanoma. J Immunother. 2014;37:348–50. doi: 10.1097/CJI.0000000000000041. [DOI] [PubMed] [Google Scholar]
  62. Voskens CJ, Goldinger SM, Loquai C, et al. The price of tumor control: an analysis of rare side effects of anti-CTLA-4 therapy in metastatic melanoma from the ipilimumab network. PLoS ONE. 2013;8:e53745. doi: 10.1371/journal.pone.0053745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Akhtari M, Waller EK, Jaye DL, Lawson DH, Ibrahim R, Papadopoulos NE, et al. Neutropenia in a patient treated with ipilimumab (anti-CTLA-4 antibody) J Immunother. 2009;32:322–4. doi: 10.1097/CJI.0b013e31819aa40b. [DOI] [PubMed] [Google Scholar]
  64. Kaehler KC, Egberts F, Lorigan P, Hauschild A. Anti-CTLA-4 therapy-related autoimmune hypophysitis in a melanoma patient. Melanoma Res. 2009;19:333–4. doi: 10.1097/CMR.0b013e32832e0bff. [DOI] [PubMed] [Google Scholar]
  65. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75. doi: 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Lipson EJ, Sharfman WH, Drake CG, et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res. 2013;19:462–8. doi: 10.1158/1078-0432.CCR-12-2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–33. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32:1020–30. doi: 10.1200/JCO.2013.53.0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Press Announcements. FDA approves Keytruda for advanced melanoma http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm412802.htm [accessed on 6 September 2014]
  70. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Ribas A, Hodi FS, Kefford R, et al. Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in 411 patients (pts) with melanoma (MEL) J Clin Oncol. 2014;32(5 Suppl):LBA9000. [Google Scholar]
  72. Hodi FS, Ribas A, Daud A, et al. Evaluation of immune-related response criteria (irRC) in patients (pts) with advanced melanoma (MEL) treated with the anti-PD-1 monoclonal antibody MK-3475. J Clin Oncol. 2014;32(5 Suppl):3006. [Google Scholar]
  73. Doukas J, Rolland A. Mechanisms of action underlying the immunotherapeutic activity of Allovectin in advanced melanoma. Cancer Gene Ther. 2012;19:811–7. doi: 10.1038/cgt.2012.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Bedikian AY, Richards J, Kharkevitch D, Atkins MB, Whitman E, Gonzalez R. A phase 2 study of high-dose Allovectin-7 in patients with advanced metastatic melanoma. Melanoma Res. 2010;20:218–26. doi: 10.1097/CMR.0b013e3283390711. [DOI] [PubMed] [Google Scholar]
  75. Liu BL, Robinson M, Han Z-Q, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10:292–303. doi: 10.1038/sj.gt.3301885. [DOI] [PubMed] [Google Scholar]
  76. Senzer NN, Kaufman HL, Amatruda T, et al. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol. 2009;27:5763–71. doi: 10.1200/JCO.2009.24.3675. [DOI] [PubMed] [Google Scholar]
  77. Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17:718–30. doi: 10.1245/s10434-009-0809-6. [DOI] [PubMed] [Google Scholar]
  78. Kaufman HL, Bines SD. OPTIM trial: a Phase III trial of an oncolytic herpes virus encoding GM-CSF for unresectable stage III or IV melanoma. Future Oncol. 2010;6:941–9. doi: 10.2217/fon.10.66. [DOI] [PubMed] [Google Scholar]
  79. Met O, Eriksen J, Svane IM. Studies on mRNA electroporation of immature and mature dendritic cells: effects on their immunogenic potential. Mol Biotechnol. 2008;40:151–60. doi: 10.1007/s12033-008-9071-6. [DOI] [PubMed] [Google Scholar]
  80. Van Brussel I, Berneman ZN, Cools N. Optimizing dendritic cell-based immunotherapy: tackling the complexity of different arms of the immune system. Mediators Inflamm. 2012;2012:690643. doi: 10.1155/2012/690643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Van Driessche A, Ponsaerts P, Van Bockstaele DR, Van Tendeloo VFI, Berneman ZN. Messenger RNA electroporation: an efficient tool in immunotherapy and stem cell research. Folia Histochem Cytobiol. 2005;43:213–6. [PubMed] [Google Scholar]
  82. Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4:328–32. doi: 10.1038/nm0398-328. [DOI] [PubMed] [Google Scholar]
  83. Aarntzen EHJG, Schreibelt G, Bol K, et al. Vaccination with mRNA-electroporated dendritic cells induces robust tumor antigen-specific CD4+ and CD8+ T cells responses in stage III and IV melanoma patients. Clin Cancer Res. 2012;18:5460–70. doi: 10.1158/1078-0432.CCR-11-3368. [DOI] [PubMed] [Google Scholar]
  84. Oshita C, Takikawa M, Kume A, et al. Dendritic cell-based vaccination in metastatic melanoma patients: phase II clinical trial. Oncol Rep. 2012;28:1131–8. doi: 10.3892/or.2012.1956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9:463–75. doi: 10.1038/nrc2656. [DOI] [PubMed] [Google Scholar]
  86. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–37. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
  87. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235:177–82. doi: 10.1126/science.3798106. [DOI] [PubMed] [Google Scholar]
  88. Molina MA, Codony-Servat J, Albanell J, Rojo F, Arribas J, Baselga J. Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Res. 2001;61:4744–9. [PubMed] [Google Scholar]
  89. Scheuer W, Friess T, Burtscher H, Bossenmaier B, Endl J, Hasmann M. Strongly enhanced antitumor activity of trastuzumab and pertuzumab combination treatment on HER2-positive human xenograft tumor models. Cancer Res. 2009;69:9330–6. doi: 10.1158/0008-5472.CAN-08-4597. [DOI] [PubMed] [Google Scholar]
  90. Mortenson ED, Fu Y-X. Adaptive immune responses and HER2/neu positive breast cancer. Curr Pathobiol Rep. 2013;1:37–42. doi: 10.1007/s40139-012-0001-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Barok M, Isola J, Pályi-Krekk Z, et al. Trastuzumab causes antibody-dependent cellular cytotoxicity-mediated growth inhibition of submacroscopic JIMT-1 breast cancer xenografts despite intrinsic drug resistance. Mol Cancer Ther. 2007;6:2065–72. doi: 10.1158/1535-7163.MCT-06-0766. [DOI] [PubMed] [Google Scholar]
  92. Perez EA, Suman VJ, Davidson NE, et al. Sequential versus concurrent trastuzumab in adjuvant chemotherapy for breast cancer. J Clin Oncol. 2011;29:4491–7. doi: 10.1200/JCO.2011.36.7045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Tagliabue E, Sfondrini L, Regondi V, et al. Chemotherapy can enhance trastuzumab-mediated ADCC. Cancer Res. 2012;72(24 Suppl) ):P5–18 – 10–P5 – 18–10. [Google Scholar]
  94. Baselga J, Cortés J, Kim S-B, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109–19. doi: 10.1056/NEJMoa1113216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Swain SM, Kim S-B, Cortés J, et al. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA study): overall survival results from a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2013;14:461–71. doi: 10.1016/S1470-2045(13)70130-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783–91. doi: 10.1056/NEJMoa1209124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Welslau M, Diéras V, Sohn J-H, et al. Patient-reported outcomes from EMILIA, a randomized phase 3 study of trastuzumab emtansine (T-DM1) versus capecitabine and lapatinib in human epidermal growth factor receptor 2-positive locally advanced or metastatic breast cancer. Cancer. 2014;120:642–51. doi: 10.1002/cncr.28465. [DOI] [PubMed] [Google Scholar]
  98. Miller KD, Diéras V, Harbeck N, et al. Phase IIa trial of trastuzumab emtansine with pertuzumab for patients with human epidermal growth factor receptor 2-positive, locally advanced, or metastatic breast cancer. J Clin Oncol. 2014;32:1437–44. doi: 10.1200/JCO.2013.52.6590. [DOI] [PubMed] [Google Scholar]
  99. Mittendorf EA, Clifton GT, Holmes JP, et al. Clinical trial results of the HER-2/neu (E75) vaccine to prevent breast cancer recurrence in high-risk patients: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02. Cancer. 2012;118:2594–602. doi: 10.1002/cncr.26574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Mittendorf EA, Storrer CE, Shriver CD, Ponniah S, Peoples GE. Investigating the combination of trastuzumab and HER2/neu peptide vaccines for the treatment of breast cancer. Ann Surg Oncol. 2006;13:1085–98. doi: 10.1245/ASO.2006.03.069. [DOI] [PubMed] [Google Scholar]
  101. Carmichael MG, Benavides LC, Holmes JP, Gates JD, Mittendorf EA, Ponniah S, et al. Results of the first phase 1 clinical trial of the HER-2/neu peptide (GP2) vaccine in disease-free breast cancer patients: United States Military Cancer Institute Clinical Trials Group Study I-04. Cancer. 2010;116:292–301. doi: 10.1002/cncr.24756. [DOI] [PubMed] [Google Scholar]
  102. Morin J, Traoré A, Dionne G, et al. Magnetic resonance-guided percutaneous cryosurgery of breast carcinoma: technique and early clinical results. Can J Surg. 2004;47:347–51. [PMC free article] [PubMed] [Google Scholar]
  103. Manenti G, Perretta T, Gaspari E, et al. Percutaneous local ablation of unifocal subclinical breast cancer: clinical experience and preliminary results of cryotherapy. Eur Radiol. 2011;21:2344–53. doi: 10.1007/s00330-011-2179-2. [DOI] [PubMed] [Google Scholar]
  104. Sabel MS, Nehs MA, Su G, Lowler KP, Ferrara JLM, Chang AE. Immunologic response to cryoablation of breast cancer. Breast Cancer Res Treat. 2005;90:97–104. doi: 10.1007/s10549-004-3289-1. [DOI] [PubMed] [Google Scholar]
  105. Sabel MS, Su G, Griffith KA, Chang AE. Rate of freeze alters the immunologic response after cryoablation of breast cancer. Ann Surg Oncol. 2010;17:1187–93. doi: 10.1245/s10434-009-0846-1. [DOI] [PubMed] [Google Scholar]
  106. Karim-Kos HE, de Vries E, Soerjomataram I, Lemmens V, Siesling S, Coebergh JWW. Recent trends of cancer in Europe: a combined approach of incidence, survival and mortality for 17 cancer sites since the 1990s. Eur J Cancer. 2008;44:1345–89. doi: 10.1016/j.ejca.2007.12.015. [DOI] [PubMed] [Google Scholar]
  107. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 2013;19:5300–9. doi: 10.1158/1078-0432.CCR-13-0143. [DOI] [PubMed] [Google Scholar]
  108. Lynch TJ, Bondarenko I, Luft A, et al. 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. 2012;30:2046–54. doi: 10.1200/JCO.2011.38.4032. [DOI] [PubMed] [Google Scholar]
  109. Reck M, Bondarenko I, Luft A, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol. 2013;24:75–83. doi: 10.1093/annonc/mds213. [DOI] [PubMed] [Google Scholar]
  110. Vlad AM, Kettel JC, Alajez NM, Carlos CA, Finn OJ. MUC1 immunobiology: from discovery to clinical applications. Adv Immunol. 2004;82:249–93. doi: 10.1016/S0065-2776(04)82006-6. [DOI] [PubMed] [Google Scholar]
  111. Sangha R, Butts C. L-BLP25: a peptide vaccine strategy in non small cell lung cancer. Clin Cancer Res. 2007;13(15 Pt 2):s4652–4. doi: 10.1158/1078-0432.CCR-07-0213. [DOI] [PubMed] [Google Scholar]
  112. Quoix E, Ramlau R, Westeel V, et al. Therapeutic vaccination with TG4010 and first-line chemotherapy in advanced non-small-cell lung cancer: a controlled phase 2B trial. Lancet Oncol. 2011;12:1125–33. doi: 10.1016/S1470-2045(11)70259-5. [DOI] [PubMed] [Google Scholar]
  113. Butts C, Socinski MA, Mitchell PL, et al. 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. 2014;15:59–68. doi: 10.1016/S1470-2045(13)70510-2. [DOI] [PubMed] [Google Scholar]
  114. McNeel D, Nguyen L, Willia E, Higano C, Paul L, Dissis M. Naturally occurring prostate cancer antigen-specific T cell responses of a Th1 phenotype can be detected in patients with prostate cancer. Prostate. 2001;47:222–9. doi: 10.1002/pros.1066. [DOI] [PubMed] [Google Scholar]
  115. Small EJ, Schellhammer PF, Higano CS, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24:3089–94. doi: 10.1200/JCO.2005.04.5252. [DOI] [PubMed] [Google Scholar]
  116. Beer TM, Schellhammer PF, Corman JM, et al. Quality of life after sipuleucel-T therapy: results from a randomized, double-blind study in patients with androgen-dependent prostate cancer. Urology. 2013;82:410–5. doi: 10.1016/j.urology.2013.04.049. [DOI] [PubMed] [Google Scholar]
  117. Kantoff PW, Schuetz TJ, Blumenstein BA, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099–105. doi: 10.1200/JCO.2009.25.0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Chow W-H, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nat Rev Urol. 2010;7:245–57. doi: 10.1038/nrurol.2010.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Rausch S, Kruck S, Stenzl A, Bedke J. IMA901 for metastatic renal cell carcinoma in the context of new approaches to immunotherapy. Future Oncol. 2014;10:937–48. doi: 10.2217/fon.14.61. [DOI] [PubMed] [Google Scholar]
  120. Thompson RH, Gillett MD, Cheville JC, et al. Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A. 2004;101:17174–9. doi: 10.1073/pnas.0406351101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Thompson RH, Kuntz SM, Leibovich BC, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res. 2006;66:3381–5. doi: 10.1158/0008-5472.CAN-05-4303. [DOI] [PubMed] [Google Scholar]
  122. Choueiri TK, Fay AP, Gray KP, et al. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann Oncol. 2014;25:2178–84. doi: 10.1093/annonc/mdu445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Thompson RH, Dong H, Lohse CM, Leibovich BC, Blute ML, Cheville JC, et al. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res. 2007;13:1757–61. doi: 10.1158/1078-0432.CCR-06-2599. [DOI] [PubMed] [Google Scholar]
  124. Kang MJ, Kim KM, Bae JS, et al. Tumor-infiltrating PD1-positive lymphocytes and FoxP3-positive regulatory T cells predict distant metastatic relapse and survival of clear cell renal cell carcinoma. Transl Oncol. 2013;6:282–9. doi: 10.1593/tlo.13256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2014 doi: 10.1200/JCO.2014.59.0703. ; doi: 10.1200/JCO.2014.59.0703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Ulloa-Montoya F, Louahed J, Dizier B, et al. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol. 2013;31:2388–95. doi: 10.1200/JCO.2012.44.3762. [DOI] [PubMed] [Google Scholar]
  127. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99. doi: 10.1056/NEJMoa1406498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Yadav M, Jhunjhunwala S, Phung QT, et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature. 2014;515:572–6. doi: 10.1038/nature14001. [DOI] [PubMed] [Google Scholar]
  129. Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–81. doi: 10.1038/nature13988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Wolchok JD, Hoos A, O'Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412–20. doi: 10.1158/1078-0432.CCR-09-1624. [DOI] [PubMed] [Google Scholar]
  131. Fojo AT, Noonan A. Why RECIST works and why it should stay. Cancer Res. 2012;72:5151–7. doi: 10.1158/0008-5472.CAN-12-0733. [DOI] [PubMed] [Google Scholar]

Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES