Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Oct 1.
Published in final edited form as: Pharmacol Ther. 2017 Mar 16;178:31–47. doi: 10.1016/j.pharmthera.2017.03.008

Current modalities in cancer immunotherapy: immunomodulatory antibodies, CARs and vaccines

Jason Lohmueller 1, Olivera J Finn 1,1
PMCID: PMC5600680  NIHMSID: NIHMS867801  PMID: 28322974

Abstract

Successes of immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cell therapy in curing patients with otherwise lethal cancers have validated immunotherapy as a treatment for cancer and have inspired excitement for its broader potential. Most promising is the ability of each approach to eliminate bulky and advanced-stage cancers and to achieve durable cures. Despite this success, to date only a subset of cancer patients and a limited number of cancer types respond to these therapies. A major goal now is to expand the types of cancer and number of patients who can be successfully treated. To this end a multitude of immunotherapies are being tested clinically in new combinations, and many new immunomodulatory antibodies and CARs are in development. A third major immunotherapeutic approach with renewed interest is cancer vaccines. While over 20 years of therapeutic cancer vaccine trials have met with limited success, these studies have laid the groundwork for the use of therapeutic vaccines in combination with other immunotherapies or alone as prophylactic cancer vaccines. Prophylactic vaccines are now poised to revolutionize cancer prevention as they have done for the prevention of infectious diseases. In this review we examine three major cancer immunotherapy modalities: immunomodulatory antibodies, CAR T cell therapy and vaccines. For each we describe the current state of the art and outline major challenges and research directions forward.

Keywords: cancer immunotherapy, cancer vaccine, immune checkpoint inhibitor, immunomodulator, chimeric antigen receptor (CAR), adoptive cell therapy

1. Introduction

Within the last two decades cancer immunotherapy, the therapeutic modulation or targeting of the immune response against cancer, has surged to the forefront of cancer research and treatment (Couzin-Frankel, 2013). Renewed interest in the field has been inspired by the dramatic success of two major cancer immunotherapies that achieve durable responses in patients with terminal stages of disease: immune checkpoint inhibitor (ICI) antibodies targeting the PD-1/CTLA-4 axes in advanced melanoma (now also in lung and renal cell carcinomas) and adoptive cell therapy (ACT) with chimeric antigen receptor (CAR) T cells targeting CD19 on B-cell leukemias and lymphomas (J. R. Brahmer et al., 2012; Brentjens et al., 2013; Brentjens et al., 2011; Hodi et al., 2010; Porter, Levine, Kalos, Bagg, & June, 2011; Topalian et al., 2012). Clinical responses to these therapies have highlighted the general power and promise of stimulating an immune response to treat cancer. Immune-based therapies can eliminate large tumor masses in advanced-stage cancer patients and elicit immunological memory that can lead to prolonged protection from cancer relapse (Chapman, D'Angelo, & Wolchok, 2015; Pedicord, Montalvo, Leiner, & Allison, 2011). These results stand in contrast to other targeted therapies that typically only extend lives by weeks and more rarely months (Maemondo et al., 2010; Maude et al., 2014; Schadendorf et al., 2015; Shaw et al., 2013). The results with CARs and ICIs validate different but complementary general therapeutic mechanisms. ICIs lead to the activation and expansion of existing tumor-specific immune cells that are otherwise suppressed in the tumor microenvironment (TME) while for CAR therapy, patients' immune cells are genetically engineered to acquire new tumor-targeting specificity and potency.

Despite the justified excitement about unprecedented clinical results, current immunotherapies are still only able to achieve durable responses in subsets of cancer patients. In the case of ICIs, only 15-25% of patients with selected tumor types (NSCLC, RCC and Merkel cell carcinoma) experience complete responses (J. Brahmer et al., 2015; Motzer et al., 2015; Nghiem et al., 2016; Postow et al., 2015). One of the biggest questions in cancer immunotherapy now is whether new ICIs or combinations of existing ICIs with other treatments can vastly improve the number of treatable patients and cancer types. To this end there has been an influx of clinical trials in which existing ICI antibodies are being combined with various standard of care therapies such as chemotherapy and radiotherapy or new immunomodulators and other immunotherapies such as vaccines (Khalil, Smith, Brentjens, & Wolchok, 2016). Currently there are >350 ongoing or planned clinical trials registered on clinicaltrials.gov in which an ICI is combined with one or more different treatments and this number is steadily growing. In the case of CAR therapy, the major success to date has been in hematologic malignancies targeting a single antigen, CD19. The predominant goal for the field now is to expand CAR therapy to additional patients and cancers by targeting other antigens including those on solid tumors (Morello, Sadelain, & Adusumilli, 2016). Successful treatment of solid tumors with CARs presents an additional challenge of creating cells that can function in the immune suppressive TME. Many CARs targeting antigens other than CD19 are already in preclinical or clinical development, and there are several therapeutic design strategies being tested to overcome TME immune suppression.

The successes of ICI and CAR therapies have led to mainstream realization that cancer is an immunological disease (Hanahan & Weinberg, 2011). It is now established that even the activity of chemotherapy and radiation depends on the patient's immune system and the ability of these standard therapies to induce immunogenic cell death (Galluzzi, Buque, Kepp, Zitvogel, & Kroemer, 2015). The idea that there is an immunological component to cancer has been posited for over 100 years beginning with the development of Coley's toxin and the work of Paul Ehrlich (Coley, 1910; Kaufmann, 2008). Observation of the abscopal effect in 1953, in which local radiation treatment of a tumor led to elimination of metastases outside the treatment area, is now appreciated to be through activation of anti-tumor immunity (Mole, 1953; Postow et al., 2012). Around this time Burnet and Thomas put forward the cancer immunosurveillance hypothesis which posits that our immune system protects us from cancer as well as from pathogens (Burnet, 1957). This hypothesis was validated with pre-clinical experiments in the 1990s, and it was more conclusively shown in the 2000s with the elucidation of the process of tumor immunoediting defined by three outcomes: tumor elimination, equilibrium and escape (Dunn, Old, & Schreiber, 2004a, 2004b). The wide acceptance of this immune function supports the development of prophylactic cancer vaccines that would strengthen or boost anti-tumor immune memory that could be reactivated in the presence of early premalignant or malignant cells leading to their elimination and cancer prevention. After over 20 years of cancer vaccines being applied in advanced-stage cancer patients, only now are the first prophylactic cancer vaccines being tested in at-risk patients. Many existing vaccines that failed in clinical trials with late-stage cancer patients are appropriate candidates for testing in the prophylactic setting (Finn & Beatty, 2016).

2. Immunomodulatory antibodies

A major shift in antibody-based immunotherapy of cancer has been in targeting immune cells in the TME instead of cancer cells. Monoclonal antibodies (mAbs) for cancer therapy have been in development since the 1970s and early efforts were focused solely on targeting tumor-associated antigens (TAAs) and directly killing tumor cells (Kohler & Milstein, 1975). Antibodies can kill target cells through a variety of mechanisms such as antibody dependent cell cytotoxicity (ADCC), complement dependent cytotoxicity, interfering with cell signaling pathways, or facilitating tumor cell phagocytosis by macrophages (Scott, Wolchok, & Old, 2012). They can also be chemically conjugated to cytotoxic drugs or radioisotopes to deliver toxic payloads, or even fused to immunomodulatory antibodies to create bi-specific molecules that target immune cells to tumor antigens. Although the development of antibodies that target TAAs is still a very promising area of investigation it is noteworthy that of the ∼20 total FDA-approved mAbs and conjugates for cancer therapies, the 5 antibodies targeting non-tumor cells in the TME were approved within the last five years (Redman, Hill, AlDeghaither, & Weiner, 2015). The first clinical application of an antibody targeting non-tumor cells in the TME targeted vascular endothelial growth factor (VEGF) affecting tumor vasculature with the goal of blocking tumor blood supply and angiogenesis, which is important for delivery of nutrients for tumor growth and metastasis (Bennouna et al., 2013). More recently immunomodulatory antibodies targeting the PD-1/PD-L1 and CTLA-4 inhibitory receptors on patients' T cells have moved to the forefront of antibody therapy showing objective responses with survival benefit in a large percentage of treated patients and complete durable responses of over 10 years for small subsets of patients (Ascierto & Marincola, 2015).

ICI antibodies function by reactivating patient's anti-tumor T cells to kill tumor cells. T cells are the major immune effector cells that mediate anti-tumor immunity, and understanding T cell activation signaling is key to understanding the activity of immunomodulatory antibodies. T cell activation is regarded as a two-signal process requiring recognition of a specific MHC-presented antigenic peptide by the T cell receptor (TCR) and a second accessory co-stimulatory signal from a co-receptor on the target cell (L. Chen & Flies, 2013). If T cells receive only the TCR signal they enter into an anergic state, become suppressive or apoptose. T cells are also susceptible to suppressive signals such as those mediated through inhibitory receptors CTLA-4 and PD-1 that engage their ligands B71/2 and PD-L1, respectively, on antigen presenting cells (APCs). Activation of these checkpoint inhibition pathways plays an important role in dampening a potentially overactive immune response when fighting pathogens in order to prevent excessive immune pathology. These same inhibitory pathways can also be detrimental to antitumor T cell responses. The ligands for CTLA-4 and PD-1 (B71/2 and PD-L1, respectively) can be co-opted by tumor cells to inhibit tumor antigen-specific T cells. Antibodies that block inhibitory receptors or their ligands on T cells, APCs, or tumor cells enhance the effectiveness of tumor-specific T cells and lead to tumor rejection (Pardoll, 2012).

With the dramatic effectiveness of this approach, a major research effort was stimulated to expand the use of these antibodies to different cancers in combinations with other immunotherapies and to develop additional immunomodulatory antibodies targeting other known T cell suppression and co-stimulation pathways. In part, this effort involves identification of patients likely to respond and identifying predictive biomarkers of response (Topalian, Taube, Anders, & Pardoll, 2016). Additionally, efforts are underway to define molecular events and therapies that would turn initial non-responders into potential responders (Gajewski, 2015).

2.1 Modulating the tumor microenvironment with immune checkpoint inhibitors: anti-CTLA-4 and anti-PD1/PD-L1 antibodies

The first T cell inhibitory receptor to be discovered was the cytotoxic T-lymphocyte associated protein 4 (CTLA-4) (Krummel & Allison, 1995; Walunas et al., 1994). As part of the process of T cell activation, CTLA-4 translocates to the cell surface and acts to limit TCR signaling by activating inhibitory signaling and by blocking the CD28 co-stimulatory receptor from binding to its ligands B7-1/2. The importance of normal immune regulation by CTLA-4 can be seen in Ctla-4 knockout mice, which develop fatal autoimmune disease with uncontrolled expansion of T cells (Waterhouse et al., 1995). CTLA-4 is also one of the key molecules used by regulatory T cells (Tregs) to suppress activation of effector T cells. In the TME it is expressed on tumor cells and on Tregs, both of which can suppress T cell lysis of tumors (Wing et al., 2008).

Allison et al. were the first to discover that antibodies blocking CTLA-4 binding to B71/2 could expand and activate anti-tumor T cells to inhibit growth of certain mouse tumors (Leach, Krummel, & Allison, 1996). This approach was tested in several Phase I and II trials leading to a Phase III validation trial in melanoma with two treatment arms – anti-CTLA-4 alone or in combination with the gp100 peptide plus adjuvant vaccine (Hodi et al., 2010). The results showed that the anti-CTLA-4 antibody improved survival even without the addition of the vaccine indicating that the antibody was relieving the suppression of preexisting tumor-specific T cells. This trial was a major success as it was the first Phase III study to show durable improvement for patients with advanced stage melanoma, increasing median survival time by 4 months. Another striking feature of the response was that patients who survived beyond 2 years continued to respond for the length of the trial suggesting a potential cure. CTLA-4 blockade has since been shown to increase the breadth of the T cell repertoire targeting the tumor instead of simply expanding a few clones (L. Robert et al., 2014). The anti-CTLA4 antibody ipilimumab received FDA approval for treatment of metastatic melanoma in 2011. Currently anti-CTLA-4 antibodies are being tested in hundreds of registered clinical trials for several cancers including bladder, prostate and lung cancer and in combinations with other therapies such as PD-1 blockade, and other immunomodulatory antibodies and small molecules as well as radiation, chemotherapy, viral therapy, vaccines, and cryoablation (Postow et al., 2012; Waitz, Fasso, & Allison, 2012; Zamarin et al., 2014).

Programmed death 1 (PD-1) was the second major inhibitory receptor discovered on T cells for which an antagonistic antibody was developed. PD-1, expressed on activated and exhausted T cells, inhibits T cell function after binding to its ligands PD-L1 and PD-L2 expressed on cancer cells and APCs. This binding acts to down-regulate T cell signaling and inhibit IL-2 production and T cell expansion. PD-L1 can also suppress T cell function by interfering with CD28-mediated co-stimulation by binding to its ligand CD80 (J. J. Park et al., 2010). Antibodies blocking either PD-1 on T cells or PD-L1 on tumor cells were therapeutic in pre-clinical mouse tumor models (Ohigashi et al., 2005; Okudaira et al., 2009). Humanized antibodies targeting PD-1 were generated (nivolumab and pembrolizumab) and showed very successful clinical results in advanced stage melanoma where 40% of patients had objective responses compared to 12% for ipilimumab (C. Robert et al., 2015). This trial led to the approval in 2014 of anti-PD-1 antibody therapy for advanced stage melanoma. Anti-PD-1 therapy has since been approved for several other cancers including NSCLC refractory to platinum therapy with a 15% overall response rate (ORR) and RCC (21.5% ORR) (J. Brahmer et al., 2015; Motzer et al., 2015). An antibody targeting PD-L1, atezolizumab, was also approved in 2016 for the treatment of bladder cancer after a Phase III trial showed a 22% ORR and is now being evaluated in trials for several other cancers (Powles et al., 2014). Importantly the combination of CTLA-4 and PD-1 ICI antibodies is also FDA-approved given an unprecedented 58% ORR in a Phase III melanoma trial including a 11.5% complete response rate (Larkin et al., 2015). The combination of these agents is considered by many to be the primary treatment modality moving forward.

While effective for many patients, it is important to note that ICI therapy can also cause serious immune-related adverse events (irAE) (Michot et al., 2016). Serious irAEs include grade III to grade IV autoimmune toxicities that are most commonly dermatologic, gastrointestinal, hepatic, or endocrine related. In a study comparing ICI treatments targeting PD-1, CTLA-4, and the combination of PD-1 and CTLA-4 for metastatic melanoma, it was found that 16.3%, 27.3%, and 55.0% of patients receiving these therapies experienced serious grade III/IV irAEs, respectively. This result indicated that the toxicities as well as the efficacies from the combined therapies appear to be additive. Higher toxicities are consistently observed for CTLA-4-targeting ICIs compared to those targeting PD-1 (T. W. Chen, Razak, Bedard, Siu, & Hansen, 2015). While most of the irAE toxicities can be managed or reversed using corticosteroids without negatively impacting tumor response, they are sometimes serious enough to warrant discontinuation of therapy (Larkin et al., 2015).

There is an ongoing effort to determine why the large majority of patients treated with anti-CTLA-4 and anti-PD-1/PD-L1 therapies do not respond. For both therapies some key biomarkers of response have emerged (Topalian et al., 2016). Expression of CTLA-4 and PD-L1 or PD-L2 pathway proteins on the tumor and/or tumor infiltrating T cells (TIL) for each respective ICI has been shown to correlate with response to therapy with the best responders being the ones with the highest levels of these molecules. The presence of a T cell infiltrate in tumors has also been shown to be important. Together these biomarkers fit with the mechanism of action of the ICIs as there must be an existing immune response that the antibodies can reactivate by blocking the immune checkpoints. Another interesting correlation with ICI response appears to be the extent of DNA mutations in the tumor (Rizvi et al., 2015; Snyder et al., 2014; Van Allen et al., 2015). This correlation suggests that the more highly mutated cancers are more likely to be under immune surveillance. However, there is no obvious threshold number of mutations and there are significant exceptions where patients with only a few mutations respond and some with highly mutated tumors do not. Looking at gene expression profiles of responders versus non-responders, type I interferon gene signature correlated with a better response (Diamond et al., 2011; Fuertes et al., 2011). It has also been demonstrated in pre-clinical studies that the gut microbiome plays a role in identifying patients who will respond to both PD-1 and CTLA-4 therapies (Sivan et al., 2015; Vetizou et al., 2015).

For patients who do not respond to PD-1 and CTLA-4 ICI antibody therapy, one major research focus is to induce conditions in the tumor that are associated with a favorable response prior to checkpoint inhibitor treatment, such as the presence of high numbers of T cells, primarily CD8+ T cells, where previously there were very few (Gajewski, 2015). Strategies include radiation, activation of innate immune signaling pathways such as stimulator of interferon genes (STING), local delivery of type I interferons, inhibition of immunosuppressive oncogene pathways such as STAT-3 and beta-catenin, and induction of tertiary lymphoid structures in the tumor (Burdelya et al., 2005; Deng et al., 2014; Oh et al., 2012; Spranger, Bao, & Gajewski, 2015). Another strategy being investigated is the identification of probiotics that will enhance therapy and induce more favorable responses as suggested by pre-clinical studies in mice that showed the importance of the gut microbiome (Iida et al., 2013). In total there are over 240 registered clinical trials investigating the effects of ICIs targeting PD-1 or PD-L1 in combination with other therapeutic modalities.

2.2 Next-generation immune checkpoint inhibitors

Immunomodulatory antibodies acting on pathways other than CTLA-4 and PD-1 are also under development. With the combination of CTLA-4 and PD-1 blockade leading to additive responses in a subset of patients, it is tempting to consider that the non-responders could be successfully treated with antibodies that suppress other inhibitory checkpoints or agonize stimulatory immune response pathways (Larkin et al., 2015). It is too early to tell whether these new therapies will lead to an incremental change in outcomes or if there will be breakthrough drugs or combinations that will significantly expand the number of treatable patients and tumor types. As the number of these agents and their potential targets grow, identification of biomarkers that can predict effectiveness of therapy will be paramount for creating rational treatment combinations. Table 1. lists and categorizes clinical trials with some of the most promising new immunomodulators that are reviewed below.

Table 1. Next-generation immunomodulatory antibodies in clinical trials.

Antibody Target Combination Conditions Phase(s) Enrollment Start Date NCT #a
4-1BB - Melanoma Phase 2 158 Mar-8 NCT00612664
4-1BB rituximab B-NHL,NSCLC,RCC,HNSCC Phase 1 220 Jun-11 NCT01307267
4-1BB anti-PD1 Advanced Solid Tumors Phase 1 45 Aug-14 NCT02179918
4-1BB anti-OX40 Neoplasms Phase 1 190 Apr-15 NCT02315066
4-1BB anti-PD-L1 Advanced Cancer Phase 1 317 Nov-15 NCT02554812
4-1BB anti-PD1, TIL therapy, cytokine Melanoma 12 Mar-16 NCT02652455
4-1BB anti-PD1 Urothelial Carcinoma,Bladder Cancer Phase 2 44 Sep-16 NCT02845323
CD27 - CD27 Expressing B-cell Malignancies, T-cell Malignancies, Solid Tumors Phase 1 90 Oct-11 NCT01460134
CD27 anti-CTLA-4, CDX-1401 Unresectable Stage III or Stage IV Melanoma Phase 1,2 100 Apr-15 NCT02413827
CD27 anti-PD-1 NSCLC, SCCHN, Ovarian
Carcinoma, Colorectal Cancer, RCC,
GBM, Melanoma
Phase 1,2 190 Jan-15 NCT02335918
CD27 anti-PD-L1 Kidney Neoplasms Phase 1,2 55 Oct-15 NCT02543645
CD27 glembatumumab vedotin Melanoma Phase 2 90 Nov-14 NCT02302339
CD27 ONT-10 vaccine Advanced Breast Carcinoma, Advanced Ovarian Carcinoma Phase 1 22 Nov-14 NCT02270372
CD27 radiotherapy Prostate Cancer Phase 1 21 Nov-14 NCT02284971
CD27 sunitinib Kidney Neoplasms Phase 1,2 58 May-15 NCT02386111
CD40 - Advanced Solid Tumors Phase 1 null Jan-4 NCT02225002
CD40 - Multiple Myeloma Phase 1 44 Mar-4 NCT00079716
CD40 - B-NHL Phase 1 50 Dec-4 NCT00103779
CD40 - CLL Phase 1 26 Apr-5 NCT00108108
CD40 - CLL Phase 1,2 12 Jul-5 NCT00283101
CD40 - MM Phase 2 33 Sep-5 NCT00231166
CD40 - B-NHL Phase 2 46 Dec-6 NCT00435916
CD40 - B-NHL Phase 1 29 Jul-7 NCT01561911
CD40 chemotherapy, dexamethasone MM Phase 1 36 Aug-7 NCT00525447
CD40 rituximab, chemotherapy B-NHL Phase 2 151 Sep-7 NCT00529503
CD40 chemotherapy, dexamethasone Neoplasms Phase 1 34 Nov-7 NCT00607048
CD40 rituximab B-NHL Phase 1 22 Dec-7 NCT00556699
CD40 - B-NHL, HL Phase 1,2 111 Mar-8 NCT00670592
CD40 rituximab, chemotherapy B-NHL Phase 1 33 Apr-8 NCT00655837
CD40 bortezomib MM Phase 1 18 May-8 NCT00664898
CD40 chemotherapy Pancreatic Neoplasm Phase 1 22 Jun-8 NCT00711191
CD40 anti-CTLA-4 Recurrent Melanoma, Stage IV Melanoma Phase 1 32 Feb-10 NCT01103635
CD40 chemotherapy Follicular Lymphoma Phase 1 1 Feb-11 NCT01275209
CD40 anti-PD-L1 Solid Cancers Phase 1 160 Dec-14 NCT02304393
CD40 - Neoplasms Phase 1 32 May-15 NCT02482168
CD40 vanucizumab Advanced/Metastatic Solid Tumors Phase 1 170 Jan-16 NCT02665416
CD40 anti-PD-1 Melanoma Phase 1,2 41 Oct-16 NCT02706353
CD73 PD-L1 Solid Tumours Phase 1 188 Jul-15 NCT02503774
GITR - Unresectable Stage III or Stage IV
Malignant Melanoma or Other Solid
Tumor Malignancies
Phase 1 40 Oct-10 NCT01239134
GITR anti-PD-1 Advanced Solid Tumor Phase 1 96 Nov-15 NCT02553499
GITR - Advanced Solid Tumors Phase 1 45 Nov-15 NCT02583165
GITR - Solid Tumors Phase 1 44 Dec-15 NCT02628574
GITR - Advanced Cancer, Metastatic Cancer Phase 1,2 146 Apr-16 NCT02697591
GITR anti-PD-1 Solid Tumors, Lymphomas Phase 1 264 Jun-16 NCT02740270
ICOS - Lymphomas Phase 1 46 16-Apr NCT02520791
ICOS anti-PD-1 Cancer Phase 1 304 Jun-16 NCT02723955
KIR anti-CTLA-4, anti-PD-1 B-NHL, HL, MM Phase 1 375 Jun-12 NCT01592370
KIR anti-PD-1 CANCER,NOS Phase 1 162 Oct-12 NCT01714739
KIR anti-CTLA-4 CANCER, NOS Phase 1 22 Dec-12 NCT01750580
KIR anti-PD-1 Solid Tumors Phase 1,2 260 Oct-15 NCT02598960
LAG-3 anti-PD-1 Neoplasms by Site Phase 1 360 Oct-13 NCT01968109
LAG-3 anti-PD-1 Hematologic Neoplasms Phase 1,2 132 Feb-14 NCT02061761
LAG-3 anti-PD-1 Advanced Solid Tumors Phase 1,2 416 Jun-15 NCT02460224
LAG-3 anti-4-1BB, anti-PD-1 Glioblastoma,Gliosarcoma,
Recurrent Brain Neoplasm
Phase 1 68 Mar-16 NCT02658981
LAG-3 anti-PD-1 Advanced Cancer Phase 1 70 May-16 NCT02720068
OX40 vaccine Advanced Cancer Phase 1 30 Nov-3 NCT01644968
OX40 chemotherapy, radiotherapy Prostate Cancer Phase 1,2 10 Oct-10 NCT01303705
OX40 radiotherapy Metastatic Breast Cancer,Lung
Metastases,Liver Metastases
Phase 1,2 40 Feb-13 NCT01862900
OX40 rituximab, anti-CTLA-4, anti-PD-L1 Advanced Solid Tumors,Aggressive
B-cell Lymphomas
Phase 1,2 58 Aug-14 NCT02205333
OX40 anti-PD-L1 Recurrent or Metastatic Solid Tumors Phase 1 39 Sep-14 NCT02221960
OX40 - Head and Neck Cancer Phase 1 55 Oct-14 NCT02274155
OX40 - Advanced Solid Tumors Phase 1 196 Mar-15 NCT02318394
OX40 anti-PD-L1, bevacizumab Solid Tumor Phase 1 762 Apr-15 NCT02410512
OX40 - Colorectal Neoplasms Phase 1 44 Sep-15 NCT02559024
OX40 anti-CTLA-4, anti-PD-L1 Select Advanced Solid Tumors Phase 1 364 Mar-16 NCT02705482
TIM-3 anti-PD-1 Advanced Malignancies Phase 1,2 250 Nov-15 NCT02608268
TIM-3 anti-PD-1 Advanced or Metastatic Solid Tumors Phase 1 402 Jul-16 NCT02817633
VISTA - Advanced Cancer Phase 1 150 Jan-16 NCT02671955
a

Trials with recruitment listed as “Completed,” “Active but not recruiting,” “Terminated” or “Suspended” on clinicaltrails.gov are italicized. Trials were obtained by bulk data download from clinicaltrials.gov on August 1st 2016.

One T cell inhibitory receptor being targeted in the clinic is T-cell membrane protein 3 (TIM-3). This receptor is up-regulated on exhausted T cells and has been shown to suppress T cell functions and induce apoptosis (Y. H. Huang et al., 2015; Sabatos et al., 2003; Zhu et al., 2005). In preclinical studies in mice with colon cancer, melanoma, and sarcoma, TIM-3 blockade in combination with PD-L1 blockade has shown a synergistic improvement in tumor elimination and control (Kim et al., 2016; Sakuishi et al., 2010). Because TIM-3 is known to interact with several ligands, such as galectin-9, HMGB1, phosphatidyl serine and CEACAM-1, the precise mechanism of TIM-3 blockade that is leading to favorable outcomes is still unclear. TIM-3 blocking antibodies are being tested in clinical trials in combination with anti-PD-1 therapy.

Another therapeutic target with an antagonistic antibody already in clinical trials is the lymphocyte-activation gene-3 (LAG-3) molecule. LAG-3 is expressed on activated effector T cells, Tregs, B cells and plasmacytoid DC (pDCs) (Baixeras et al., 1992). Mechanistically, LAG-3 acts as a direct competitor for MHC-II binding to CD4, which leads to suppression of T cell activation and increase of the suppressive abilities of Tregs (C. T. Huang et al., 2004). Anti-LAG-3 antibodies work to block this interaction. LAG-3 is co-expressed with PD-1 on exhausted T cells and the two act synergistically in tumor models (Blackburn et al., 2009; Woo et al., 2012). Thus, efforts are focused on simultaneously blocking the effect of both molecules. Several trials are underway testing LAG-3 and PD-1 dual blockade in a variety of solid tumors as well as one trial in combination with an anti-4-1BB agonist antibody.

KIR2DL1/2L3 is an inhibitory member of the Killer-cell immunoglobulin-like receptor (KIR) family of receptors found on NK cells and some T cells. Binding to MHC Class I leads to inhibition of NK-mediated cell killing, and antibodies targeting this molecule have been shown in pre-clinical work to reactivate this NK function. Multiple trials are underway testing the anti-KIR antibody lirilumab in combination with other ICIs as well as TAA-targeting antibodies believed to act via NK-mediated ADCC (Kohrt et al., 2014; Romagne et al., 2009).

Yet another ICI antibody in clinical trials targets V-domain Ig suppressor of T cell activation (VISTA). VISTA is found on hematopoeitic cells including T cells and DCs as well as on tumor cells. VISTA was shown to inhibit T cell proliferation and cytokine production in vitro as well as anti-tumor immunity in pre-clinical mouse experiments. Pre-clinical experiments show that treatment with a VISTA-specific antagonist antibody relieves this inhibition and controls tumor growth in several mouse tumor models. The anti-VISTA ICI has also been shown to synergize with PD-1 blockade (Le Mercier et al., 2014; Lines et al., 2014; L. Wang et al., 2011).

A complementary strategy to inhibiting immune checkpoint pathways is to instead target co-stimulatory receptors with agonistic antibodies. Several of these antibodies are currently in clinical trials, and antibodies targeting molecules such as CD40, OX40 and 4-1BB have been in trials since the mid-late 2000s (Table 1.). The main advance in the new trials has been targeting these molecules in combination with other therapies based on extensive pre-clinical work showing synergy with other therapeutic modalities.

One co-stimulatory target protein is the 4-1BB receptor that is expressed on T cells, NK cells and monocytes (Shuford et al., 1997). Binding of 4-1BB on T cells to its ligand 4-1BBL on DCs leads to enhanced proliferation and up-regulation of anti-apoptotic proteins, ultimately protecting T cells from activation induced cell death (AICD) (H. W. Lee et al., 2002; Starck, Scholz, Dorken, & Daniel, 2005). Pre-clinical studies have been carried out investigating the effects of anti-4-1BB agonist therapy alone and in combination with several other modalities and found it to improve tumor regression (May, Chen, Zheng, & Liu, 2002; Melero et al., 1997). While initial clinical testing of anti-4-1BB was halted due to liver toxicity, there are new ongoing trials testing it in combination with various drugs (Bartkowiak et al., 2015; Kocak et al., 2006; A. Yonezawa, Dutt, Chester, Kim, & Kohrt, 2015).

OX40 is a co-stimulatory receptor expressed on T cells, NK cells and neutrophils. It interacts with OX40L found on APCs and various other immune cells (Linch, McNamara, & Redmond, 2015). Activation of OX40 leads to T cell proliferation, survival and cytokine secretion and was also found to induce AICD in Tregs (Hirschhorn-Cymerman et al., 2009; Rogers, Song, Gramaglia, Killeen, & Croft, 2001). Anti-OX40 antibodies have been shown to mediate T cell-dependent anti-tumor activity and establishment of tumor-specific memory in pre-clinical experiments in mice (Pan, Zang, Weber, Meseck, & Chen, 2002). Anti-tumor activity was recently observed in a Phase I clinical trial for melanoma and RCC, and other trials are ongoing (Curti et al., 2013).

Glucocorticoid-induced TNFR-related protein (GITR) is another co-stimulatory receptor up-regulated on activated T cells and constitutively expressed on Tregs (Nocentini & Riccardi, 2005). Unlike the similarly regulated immune checkpoint proteins, GITR acts to enhance function of effector T cells and confers resistance to the inhibitory effects of Tregs (Kanamaru et al., 2004; Ronchetti et al., 2007). In pre-clinical studies, agonistic GITR antibodies mediated tumor rejection in several mouse cancer models (Cohen et al., 2010; Ko et al., 2005; Schaer et al., 2013). Anti-GITR antibodies are currently in early phase clinical trials for several different cancers.

CD40 is normally expressed on macrophages, monocytes and B cells, as well as on various cancers including melanoma, lymphoma, leukemia and carcinomas. The ligand for CD40, CD40L, is expressed on CD4+ T cells and plays a role in the activation of T cells by APCs (Eliopoulos & Young, 2004). There is promising pre-clinical data for the effects of anti-CD40 agonistic antibodies that are being tested in several clinical trials (Hamzah et al., 2008; Horton et al., 2010; Luheshi et al., 2016; White et al., 2011). Currently the most promising results have been with anti-CD40 antibodies combined with chemotherapy to treat pancreatic cancer (G. L. Beatty et al., 2011). Unexpectedly, in mouse models the predominant anti-tumor response appears to depend on anti-tumor effects of activated macrophages.

Another new target being explored is the CD73 ecto-5′-nucleotidase that leads to the degradation of AMP into adenosine, a small molecule metabolite that promotes immunosuppression and angiogenesis in the TME (Antonioli, Yegutkin, Pacher, Blandizzi, & Hasko, 2016). It is found primarily on Tregs and is a major contributor to their immunosuppressive activity. It is also found on tumor cells and other stromal cells in the TME. Pre-clinical models with antibodies targeting CD73 alone or in combination with other modalities such as anti-CTLA-4 and anti-PD-1, have shown significant anti-tumor activity (Allard, Pommey, Smyth, & Stagg, 2013; Loi et al., 2014). Finally, a trial looking at anti-CD73 monotherapy or combined therapy with PD-L1 ICI therapy is underway.

CD27 is a co-stimulatory molecule in the TNF-super family related to CD40, 4-1BB and OX40 that is constitutively expressed on T cells and some B and NK cells. Its only known ligand is CD70. Activation of CD27 leads to activation of the NF-kB pathway and cell survival, activation and proliferation. CD27 activation has also been shown to promote broadening of the T cell repertoire. Anti-CD27 agonist antibodies have shown activity in pre-clinical models enhancing CD8+ T cell responses and having a synergistic effect with anti-PD-1 therapy. Clinically the agonist antibody varlilumab has been shown to be well-tolerated and to have several positive effects in subsets of patients including decreasing the number of Tregs and mediating CD8+ T cell responses. Interestingly, varlilumab can also act to directly kill tumor cells that are over-expressing CD27 by mediating ADCC (He et al., 2013; Thomas, He, Marsh, & Keler, 2014).

An additional co-stimulatory molecule targeted clinically by an immunomodulatory antibody is inducible T-cell co-stimulator (ICOS) (Hutloff et al., 1999). ICOS is expressed on activated T cells and is especially important for Th2 T cell responses. Pre-clinical experiments showed that ICOS knockout dramatically decreases the efficacy of anti-CTLA-4 treatment in controlling and eliminating tumors. Agonistic anti-ICOS antibody can synergize with CTLA-4 blockade in the anti-tumor effect (Fan, Quezada, Sepulveda, Sharma, & Allison, 2014).

3. CAR T cell therapy

CARs are engineered antigen receptor proteins consisting of an antigen binding region and TCR signaling domains. When expressed on a patient's T cells, CARs act to re-direct T cells' effector functions, target cell lysis and cytokine production upon binding to antigens on tumor cells (Sadelain, Brentjens, & Riviere, 2013). T cells are genetically modified, most commonly via a retrovirus or a lentivirus that encodes the CAR, expanded and then adoptively transferred back into the patient (Barrett, Singh, Porter, Grupp, & June, 2014).

The major clinical success of CAR T cell therapy so far has been with CARs targeting CD19 in the treatment of refractory pediatric and adult B-ALL. Across multiple trials conducted by various groups, CD19-CAR therapy has resulted in astounding ∼70-95% complete response rates compared to an expected 7% 5-year survival rate for standard therapy for refractory adult ALL (Davila et al., 2014; Fielding et al., 2007; Kebriaei et al., 2016; D. W. Lee et al., 2014; D. W. Lee et al., 2015; Maude et al., 2014; J. H. Park et al., 2014). CD19-targeting CARs have also shown modest success in treating adult chronic lymphoblastic leukemia (CLL) (28% complete response rate) and are showing early promise in clinical trials for other hematological malignancies including B-NHL and MM (Garfall et al., 2015; Kochenderfer et al., 2015; J. H. Park et al., 2014; Savoldo et al., 2011; Schuster et al., 2014). The first CARs were made as early as 1989, however it took 22 years to attain this first clinical success with CAR therapy (Gross, Waks, & Eshhar, 1989). This success was the culmination of several technological advancements including those that facilitate gene delivery, enhance T cell growth and expansion, as well as CAR design considerations including the choice of CD19 antigen and most importantly the addition of co-stimulatory signaling domains such as CD28 and 4-1BB that led to enhanced CAR T cell persistence, proliferation and potency (Maher, Brentjens, Gunset, Riviere, & Sadelain, 2002; Zhong, Matsushita, Plotkin, Riviere, & Sadelain, 2010).

In addition to further optimizing CD19-CAR therapy to treat other CD19+ malignancies, a major goal in the CAR therapy field is to select new tumor antigens for a broader range of cancers, including additional hematological malignancies and solid tumors (Morello et al., 2016). This research is still focused on the overarching issues of antigen choice, CAR T cell persistence in vivo, proliferation and potency, although the TME of solid cancers poses additional engineering challenges. In response, researchers are using new design strategies that are being tested both pre-clinically and/or in clinical trials. Table 2. contains a list of registered clinical trials using CAR T cells organized by the different antigens that they target.

Table 2. CAR clinical trials by target antigen.

Antigen Signaling and
accessory
Cell
typea
Delivery
methodb
Disease(s) Primary
institution(s)
Phase(s) Enrollment NCT #c
BCMA 41BBz T cells NA MM UPenn Phase 0 30 NCT02546167
BCMA 41BBz T cells Lenti MM bluebird bio Phase 1 50 NCT02658929
BCMA 41BBz T cells Lenti MM bluebird bio 50 NCT02786511
CD7 CD28-41BBz NK-92 NA AML PersonGen Phase1,2 10 NCT02742727
CD19 41BBz T cells Retro B-ALL, B-CLL, B-NHL UPenn 110 NCT01029366
CD19 41BBz T cells (allo) Lenti B-ALL UPenn Phase 1 10 NCT01551043
CD19 41BBz T cells Lenti B-ALL, B-CLL, B-NHL UPenn Phase 1 20 NCT01626495
CD19 41BBz T cells Lenti B-CLL, B-SLL UPenn Phase 2 65 NCT01747486
CD19 41BBz T cells 1:1 CD4+ and CD8+ Lenti B-ALL Seattle Children's Phase1,2 80 NCT02028455
CD19 41BBz T cells Lenti B-NHL UPenn Phase 2 51 NCT02030834
CD19 41BBz T cells Lenti B-ALL UPenn Phase 2 24 NCT02030847
CD19 41BBz T cells RNA (electro) HL UPenn Phase 0 16 NCT02277522
CD19 41BBz T cells Lenti B-NHL Novartis Phase 2 100 NCT02445248
CD19 41BBz T cells RNA (electro) HL UPenn|CHOP Phase 0 10 NCT02624258
CD19 41BBz T cells Lenti B-CLL, B-SLL UPenn 15 NCT02640209
CD19 41BBz T cells Lenti B-NHL UPenn Phase1,2 12 NCT02650999
CD19 41BBz T cells Lenti B-CLL Shanghai GeneChem Phase1,2 30 NCT02672501
CD19 41BBz T cells Lenti MM UPenn Phase 2 25 NCT02794246
CD19 41BBz and zeta-only T cells Retro B-ALL, B-CLL, B-NHL Chinese PLAGH 12 NCT01864889
CD19 41BBz and zeta- only T cells Retro B-NHL Chinese PLAGH Phase1,2 2 NCT02081937
CD19 41BBz,CD20/34t T cells (allo, gen mod) Lenti B-ALL, B-CLL, B-NHL Servier 200 NCT02735083
CD19 41BBz,CD20/34t T cells (allo, gen mod) Lenti B-ALL, B-CLL Servier Phase 1 12 NCT02746952
CD19 41BBz,EGFRt T cells Lenti B-ALL, B-CLL, B-NHL FHCRC|NCI Phase1,2 145 NCT01865617
CD19 41BBz,EGFRt T cells Lenti B-NHL Juno Phase 1 70 NCT02631044
CD19 41BBz,EGFRt T cells Lenti B-NHL FHCRC|NCI|Juno MedImmune AstraZeneca Phase 1 42 NCT02706405
CD19 CD27z,iCasp9 T cells Lenti B-NHL PKU|UF Phase1,2 20 NCT02247609
CD19 CD28-41BBz T cells Retro B-CLL, B-NHL Uppsala Univ. Phase1,2 15 NCT02132624
CD19 CD28-41BBz,EGFRt CMV or EBV Tcm cells (allo) NA B-ALL, B-CLL, B-NHL FHCRC\\NCI Phase1,2 1 NCT01475058
CD19 CD28z T cells Retro B-NHL NCI\NIHCC Phase 1 43 NCT00924326
CD19 CD28z T cells NA B-ALL MSKCC Phase 1 60 NCT01044069
CD19 CD28z T cells (allo) Virus B-ALL, B-CLL, B cell lymphomas NCI\NIHCC Phase 1 42 NCT01087294
CD19 CD28z T Cells Transpos (electro) B-NHL M.D. Anderson Intrexon|Ziopharm Phase 1 60 NCT00968760
CD19 CD28z EBV-CTLs (allo) NA B-ALL, B-NHL MSKCC Phase 1 12 NCT01430390
CD19 CD28z T Cells (allo) Transposon (electro) B-ALL, B-CLL, B-NHL M.D. Anderson Intrexon|Ziopharm Phase 1 140 NCT01497184
CD19 CD28z T cells Retro B-ALL, B-NHL NCI\NIHCC Phase 1 52 NCT01593696
CD19 CD28z T cells NA B-ALL, B-CLL, B-NHL M.D. Anderson ZiopharmIIntrexon Phase 1 30 NCT02529813
CD19 CD28z T cells NA B-NHL MSKCC Phase 1 17 NCT01840566
CD19 CD28z T cells NA B-ALL MSKCC|Dana-Farber Phase 1 24 NCT01860937
CD19 CD28z T cells (allo) Retro B-ALL, B-CLL, B-NHL Baylor Phase 1 40 NCT02050347
CD19 CD28z T cells Retro B-NHL Jichi Med. Univ.|Takara Phase1,2 18 NCT02134262
CD19 CD28z T cells Retro B-NHL Kite Pharma Phase1,2 124 NCT02348216
CD19 CD28z T cells Retro B-ALL Juno Phase 2 90 NCT02535364
CD19 CD28z T cells Retro B-NHL Kite Pharma Phase 2 70 NCT02601313
CD19 CD28z T cells Retro B-ALL Kite Pharma Phase1,2 75 NCT02614066
CD19 CD28z T cells Retro B-ALL Kite Pharma Phase1,2 75 NCT02625480
CD19 CD28z T cells Retro B-NHL Xuzhou Med. College Phase1,2 20 NCT02652910
CD19 CD28z T cells Retro B-ALL Juno 500 NCT02813252
CD19 CD28z T cells Retro B-ALL, B-NHL Sheba Med. Center Phase1,2 40 NCT02772198
CD19 CD28z and 41BBz T cells Lenti B-ALL AMMS|PKU Phase 1 5 NCT02186860
CD19 CD28z and 41BBz T cells NA B-ALL, B-CLL, B-NHL Xuzhou Med. College Phase1,2 20 NCT02685670
CD19 CD28z and CD28-41BBz T cells Retro B-ALL, B-CLL, B-NHL Baylor Phase 1 14 NCT01853631
CD19 CD28z and zeta-only Tcm-enriched T cells Lenti B-NHL City of Hope|NCI Phase1,2 57 NCT01318317
CD19 CD28z,EGFRt T cells Lenti B-ALL Seattle Children's Phase 1 18 NCT01683279
CD19 CD28z,EGFRt T cells Tcm or Tn,Tnem Lenti B-ALL City of Hope|NCI Phase 1 48 NCT02146924
CD19 CD28z,undefined safety switch T cells Retro or Lenti B-ALL, B-CLL, B-NHL Shenzhen Second People's Hospital Phase 1 36 NCT02456350
CD19 NA T cells NA B-ALL, B-CLL, B-NHL Southwest Hospital, China Phase1,2 45 NCT02349698
CD19 NA T cells NA B-ALL, B-CLL, B-NHL ICT 30 NCT02813837
CD19 NA T cells Lenti B-ALL, B-CLL, B-NHL Tongji Univ. Med. Phase1,2 40 NCT02537977
CD19 NA T cells NA B-CLL SMMU Phase1,2 20 NCT02644655
CD19 NA T cells (allo) NA B-ALL AMMS|Chinese PLAGH Phase 1 10 NCT02799550
CD19 NA T cells NA B-NHL Sinobioway Cell Therapy Phase1,2 24 NCT02728882
CD19 NA T cells NA B-ALL Sinobioway Cell Therapy Phase1,2 24 NCT02735291
CD19 NA T cells NA B-NHL Univ. College, London Phase 1 12 NCT02431988
CD19 NA T cells NA B-ALL, B-CLL, B-NHL Beijing Doing Biomed. Phase 1 100 NCT02546739
CD19 NA YδT-cells (allo) NA B-ALL, B-CLL, B-NHL Beijing Doing Biomed. Phase 1 48 NCT02656147
CD19 NA T cells Retro B-ALL, B-CLL, B-NHL NCI|NIHCC Phase 1 64 NCT02659943
CD19 NA T cells NA B-ALL Guangdong General Hospital|CAS Phase 1 30 NCT02822326
CD19 NA T cells NA B-ALL, B-NHL Univ. College, London Phase 1 18 NCT02443831
CD19 NA T cells NA B-CLL iCarTAB Xuzhou Med. Phase1,2 50 NCT02782351
CD19 NA T cells NA B-NHL PKU|Marino Phase 1 20 NCT02842138
CD19 NA T cells NA B-ALL, B-CLL, B-NHL PersonGen Phase1,2 10 NCT02851589
CD19 NA T cells NA B-ALL, B-CLL, B-NHL PersonGen Phase1,2 10 NCT02819583
CD19 zeta-only T cells Retro B-NHL Christie NHS Foundation Phase 1 24 NCT01493453
CD19, CD20 NA T cells NA B-NHL Southwest Hospital, China Phase1,2 40 NCT02737085
CD19, Mesothelin 41BBz T cells Lenti Pancreatic Cancer UPenn\UCSF Phase 1 12 NCT02465983
CD20 NA T cells Virus B-ALL, B-CLL, B-NHL Southwest Hospital, China Phase1,2 45 NCT02710149
CD22 41BBz T cells Lenti B-ALL, B-NHL NCI|NIHCC Phase 1 57 NCT02315612
CD22 41BBz T cells Lenti B-ALL UPenn 15 NCT02588456
CD22 41BBz T cells Lenti B-ALL UPenn|CHOP Phase 1 15 NCT02650414
CD22 41BBz T cells Retro B-NHL Xuzhou Med. Phase 1 20 NCT02721407
CD22 NA T cells Virus B-ALL, B-CLL, B-NHL iCarTAB|Xuzhou Med. Phase1,2 10 NCT02794961
CD30 CD27z,iCasp9 T cells Lenti HL, B-NHL PKU|UF Phase1,2 20 NCT02274584
CD30 CD28z T cells Virus HL, B-NHL UNC Lineberger|Baylor Phase 1 18 NCT01316146
CD30 CD28z T cells Virus HL, B-NHL UNC Lineberger Phase 1 18 NCT02663297
CD30 CD28z T cells Virus HL, B-NHL UNC Lineberger Phase1,2 31 NCT02690545
CD30 NA T cells NA HL, B-NHL Chinese PLAGH Phase1,2 30 NCT02259556
CD33 41BBz and zeta-only T cells Retro AML Chinese Phase1,2 10 NCT01864902
CD33 41BBz and zeta-only T cells Retro AML AMMS|Chinese PLAGH Phase 1 12 NCT02799680
CD70 NA T cells Retro CD70+ cancers NCI|NIHCC Phase1,2 113 NCT02830724
CD123 41BBz T cells RNA (electro) AML UPenn Phase 0 7 NCT02623582
CD123 CD28z,EGFRt T cells Lenti AML City of Hope NCI Phase 1 30 NCT02159495
CD133 41BBz and zeta-only T cells Retro AML, B-ALL, various solid tumors Chinese PLAGH Phase 1 20 NCT02541370
CD138 41BBz and zeta-only T cells Retro MM Chinese PLAGH Phase1,2 10 NCT01886976
CD171 CD28-41BBz,EGFRt and 41BBz,EGFRt T cells 1:1 CD4+ and CD8+ Lenti Neuroblastoma Seattle Children's Phase 1 80 NCT02311621
CEA CD28z T cells NA Various solid tumors Roger Williams Phase 2 48 NCT01723306
CEA NA T cells NA Various solid tumors Southwest Hospital, China Phase 1 75 NCT02349724
CEA zeta-only T cells Retro Various solid tumors Cancer Research UK Phase 1 14 NCT01212887
EGFR 41BBz and zeta-only T cells Lenti EGFR+ Solid Tumors Chinese PLAGH Phase1,2 60 NCT01869166
EGFRvIII 41BBz T cells Lenti Glioma UPenn|UCSF 12 NCT02209376
EGFRvIII 41BBz T cells Lenti Glioblastoma UPenn 8 NCT02666248
EGFRvIII CD28-41BBz T cells Retro Glioma NCI|NIHCC Phase1,2 18 NCT01454596
EGFRvIII NA T cells Lenti Glioma RenJi Hospital Phase 1 10 NCT02331693
EGFRvIII NA T cells Retro Glioblastoma Duke Univ. Phase 1 48 NCT02664363
EGFRvIII NA,EGFRt T cells Lenti Glioblastoma Beijing Sanbo Marino Phase 1 20 NCT02844062
EPCAM NA T cells NA Stomach Neoplasms Sinobioway Phase1,2 19 NCT02725125
EPCAM NA T cells NA Liver Neoplasms Sinobioway Phase1,2 25 NCT02729493
EPHA2 NA T cells NA Glioma Fuda, Guangzhou Phase1,2 60 NCT02575261
ErbB dimers CD28z,4aβ T cells Retro Head and Neck Cancer King's College Phase 1 30 NCT01818323
FAP CD28z CD8+ T cells Retro Mesothelioma Univ. of Zurich Phase 1 6 NCT01722149
GD2 CD28-41BB-CD27z,iCasp9 T cells Lenti Neuroblastoma Zhujiang Hospital Phase 2 30 NCT02765243
GD2 CD28- T cells Retro Neuroblastoma Baylor|NCI Phase 1 11 NCT01822652
GD2 CD28-OX40z,iCasp9 NK T cells Retro Sarcoma, Osteosarcoma, Neuroblastoma, Melanoma NCI|NIHCC Phase 1 72 NCT02107963
GD2 CD28-OX40z,iCasp9 VZV T cells Retro Sarcomas Baylor|NCI Phase 1 26 NCT01953900
GD2 CD28-OX40z,iCasp9 NK T cells Retro Neuroblastoma Baylor Phase 1 18 NCT02439788
GD2 CD28z T cells Virus Neuroblastoma Cancer Research UK Phase 1 27 NCT02761915
GD2 NA triVirus-CTLs (allo) Retro Neuroblastoma Children's Mercy, KC Phase 1 5 NCT01460901
GPC3 41BBz T cells Virus Hepatocellular Carcinoma Shanghai GeneChem Phase1,2 30 NCT02715362
GPC3 CD28-41BBz T cells Virus Hepatocellular Carcinoma RenJi Hospital Phase 1 20 NCT02395250
GPC3 NA T cells NA Hepatocellular Carcinoma Fuda, Guangzhou Phase1,2 60 NCT02723942
HER2 41BBz and zeta-only T cells Virus HER-2+ Solid Tumors Chinese PLAGH Phase1,2 10 NCT01935843
HER2 CD28z T cells Virus Sarcoma Baylor Phase 1 36 NCT00902044
HER2 CD28z CMV T cells Virus Glioblastoma Baylor Phase 1 16 NCT01109095
HER2 CD28z T cells Retro Breast Cancer Fuda, Guangzhou Phase1,2 60 NCT02547961
HER2 CD28z T cells Virus Glioblastoma Baylor Phase 1 14 NCT02442297
HER2 CD28z,TGFβDN EBV T cells Retro HER2+ Malignancies Baylor Phase 1 19 NCT00889954
HER2 NA T cells NA Various solid tumors Southwest Hospital, China Phase1,2 60 NCT02713984
IL-1-RAP NA NA NA CML Univ. Hospital of Besancon 40 NCT02842320
IL-13R 41BBz,CD19t T cells Tcm-enriched Lenti Glioma City of Hope|NCI Phase 1 75 NCT02208362
Kappa CD28z T cells Retro B-CLL, B-NHL Baylor Phase 1 54 NCT00881920
LeY CD28z T cells Retro MM, AML, MDS Peter MacCallum Phase 1 6 NCT01716364
Mesothelin 41BBz T cells RNA (electro) Mesothelioma Upenn Phase 1 18 NCT01355965
Mesothelin 41BBz T cells Lenti Various solid tumors UPenn Phase 1 21 NCT02159716
Mesothelin 41BBz T cells Lenti Various solid tumors UPenn 50 NCT02388828
Mesothelin 41BBz T cells Retro Various Mesothelin+ Tumors Chinese PLAGH Phase 1 20 NCT02580747
Mesothelin 41BBz T cells Virus Pancreatic Cancer Shanghai GeneChem Phase 1 30 NCT02706782
Mesothelin CD28-41BBz T cells Retro Various solid tumors NCI|NIHCC Phase1,2 15 NCT01583686
Mesothelin CD28z-iCasp9 T cells Virus Mesothelioma, Lung and Breast Cancers MSKCC Phase 1 24 NCT02414269
Mesothelin NA T cells Virus Breast Cancer MSKCC Phase 1 24 NCT02792114
MUC1 CD28-41BBz T cells Virus Various solid tumors PersonGen Phase1,2 20 NCT02587689
MUC1 CD28-41BBz T cells Virus Glioma, colorectal, gastric PersonGen Phase1,2 20 NCT02617134
MUC1 NA pNK cells NA Various solid tumors PersonGen Phase1,2 10 NCT02839954
MUC16 CD28zfIL-12 T cells NA Ovarian Cancer MSKCC Phase 1 30 NCT02498912
NKG2D NA T cells Virus AML, Myelodysplastic Syndrome, MM Celdara Med. Phase 1 21 NCT02203825
PSCA zeta-only, iCD40/MyD88 T cells Retro Pancreatic Cancer Bellicum Phase 1 30 NCT02744287
PSMA CD28z T cells Retro Prostate Cancer Roger Williams Phase 1 18 NCT00664196
PSMA CD28z T cells Virus Prostate Cancer MSKCC Phase 1 18 NCT01140373
ROR1 41BBz T cells NA B-ALL, B-CLL, B-NHL, Various solid tumors FHCRC|NCI Phase 1 60 NCT02706392
a

Unless specified cells are autologous patient-derived cells.

b

”Virus” indicates that the trial description mentions introducing the CAR by viral transduction but does not specify the kind of virus used.

c

Trials with recruitment listed as “Completed,” “Active but not recruiting,” “Terminated” or “Suspended” on clinicaltrails.gov are italicized. Trials were obtained by bulk data download from clinicaltrials.gov on August 1st 2016.

3.1 CAR-targeted antigens: beyond CD19

The focus on CD19 was paramount to the CAR therapy success, as CD19 has many ideal characteristics of a prototype antigen. It is present on nearly all cancer cells within a patient, thus targeting it is likely to eliminate the cancer; it plays an active role in tumorigenesis so cancer cells are less likely to escape; and it is specific to cancer cells and non-essential tissues. While the downside of the incomplete tumor specificity is that tumor rejection is accompanied with deletion of normal cells in the B cell lineage, this toxicity is manageable clinically and has been well tolerated by patients when treated with regular intravenous immunoglobulin (Davila et al., 2014; Maude et al., 2014).

The tumor-specificity of a CAR is especially important to consider. Clinical trials with adoptively transferred T cells expressing a MAGE-A3 targeted affinity-enhanced TCR and CAR cells targeting ErbB2 resulted in lethal on-target/off-tumor toxicities (Cameron et al., 2013; Morgan et al., 2010). There are currently over 30 tumor antigens being targeted by CARs in registered clinical trials and even more are in pre-clinical development. There are very few antigens that are either entirely tumor-specific or specific to tumors and non-essential tissues, like CD19. For hematological cancers these antigens include CD22 and CD20 (Haso et al., 2013; Till et al., 2012). For solid tumors they include abnormally glycosylated MUC1 and CSPG4 proteins and the abnormal splice variant of EGFR, EGFRvIII (Beard et al., 2014; Johnson et al., 2015; Lohmueller et al., 2016; Wilkie et al., 2008). Search for other abnormally glycosylated or abnormally spliced targets could yield exciting new antigen targets for CAR therapy. The great majority of tumor targets being tested show over-expression on tumors, and lower levels of expression on normal tissues (hTERT, CEA, PSMA, GD2, MUC16, HER2/ERBB2, MSLN, IL-13R, alphaFR, EpCAM, RORgamma, CD4, CD33, FR, kappa-light chain and VEGFR-2). For each of these antigens, extensive safety testing will be required and potentially a transient CAR T treatment as in the case of MSLN targeted CAR (G. L. Beatty et al., 2014). Most CAR clinical trials are set up as dose-escalation studies testing a wide range of T cell numbers. It has been observed that lower numbers of potentially harmful CAR T cells show less toxicity, indicating that perhaps off-target toxicity can be managed by transferring fewer cells. However, none of these trials so far has shown a durable anti-tumor response. Alternative methods being explored to lower toxicity are limiting the CAR T cell activity to the tumor by either intratumoral injection and/or local activation or by affinity-tuning the antigen binding domain of the CAR (Adusumilli et al., 2014; Liu et al., 2015; Roybal et al., 2016).

In addition to tumor specificity there are other major considerations when choosing CAR targets. Many of these characteristics were emphasized in the report from the 2009 NCI workshop in which 75 tumor antigens were systematically evaluated and prioritized as candidates for cancer vaccines (Cheever et al., 2009). This prioritization was based on several criteria including proven therapeutic function, immunogenicity, role in oncogenicity, specificity, expression level, expression on cancer stem cells, the number of patients with tumors expressing the antigen, the number of epitopes and cellular localization. Similar criteria apply to CAR antigens and many of these antigens could be prioritized for targeting by CARs.

Another exciting approach that effectively increases the tumor-specificity of CAR therapy is based on antigen combinations using logic gate CARs. The first of these gates demonstrated was a logical “AND gate” for which only cells expressing antigens A AND B are lysed by CAR cells (Kloss, Condomines, Cartellieri, Bachmann, & Sadelain, 2013). This behavior was accomplished by creating a 1st generation CD3-zeta CAR with a weakened antigen binding domain for one target antigen, co-expressed with a co-stimulatory CD28 CAR recognizing the second antigen. Another logical circuit designed was an A AND NOT B circuit in which cells with antigen A but not antigen B were killed, using an inhibitory CAR to inhibit CAR signaling in response to antigen B (Fedorov, Themeli, & Sadelain, 2013). A more recent approach uses a receptor to sense the first signal and then turn on the production of a CAR to sense the second signal (Roybal et al., 2016). Finally, while most CARs target tumor-expressed antigens, there are also emerging examples of CARs targeting molecules specific to immunosuppressive cells in the TME. In 2014 Wang et al. created a CAR to target fibroblast activation protein (FAP) expressed on cancer-associated fibroblasts (CAF), leading to better tumor control without major toxicities (L. C. Wang et al., 2014). The recent case report of CD19 CAR T cells leading to a complete remission of a CD19- cancer suggests that the CD19 CAR could be functioning not only by destroying CD19+ tumor cells but possibly also by eliminating CD19+ immunosuppressive regulatory B cells (Garfall et al., 2015).

3.2 Optimizing CAR design and therapy

Further work is being applied to optimize the CAR protein design as well as other parts of CAR therapy including immunodepletion pre-conditioning therapy, using different defined T cell subtypes as therapeutic cells and finally the addition of accessory proteins to augment the CAR T cells' activity. All parts of the CAR, the antigen binding region, extracellular spacer, transmembrane domain, co-signaling domain and activation domain, have undergone some level of design optimization (Sadelain et al., 2013). From both pre-clinical and clinical studies principles of CAR design have emerged, although aspects of each are still active areas of investigation. Ideally one could imagine the creation of a universal “best CAR” architecture that could be used interchangeably with any new antigen binding region. However, it is unclear whether such an architecture is possible. For one, it has been shown that the immune synapse, the space between the T cell and target cell, is important for CAR efficiency, and the extracellular spacer length for each new antigen targeted might need to be tailored to that antigen (Haso et al., 2013; Hudecek et al., 2015). It is also possible that certain co-signaling domain configurations will be better suited to different TMEs requiring personalized CAR architectures for individual patients, similar to ICI antibody combinations (Condomines et al., 2015). Co-signaling domains to date include CD28, 4-1BB, ICOS, DAP10 and OX40 (Altvater, Pscherer, Juergens, & Rossig, 2005; Guedan et al., 2014; Maher et al., 2002; Zhong et al., 2010). Each co-stimulatory domain drives distinct biomolecular pathways leading to unique effects on CAR persistence, survival, metabolism and T cell fate (Kawalekar et al., 2016; Long et al., 2015). When over-stimulated these co-receptors can also have negative effects such as causing AICD and T cell exhaustion (Hombach & Abken, 2011). Interestingly, in the case of CD19 CAR trials, several different architectures including different extracellular spacer domains and co-stimulatory domains have had very similar clinical outcomes. These results suggest that for treating some cancers the specific CAR architecture does not have a major impact on outcome. So far the CARs in clinical testing have been developed through rational empirical testing of between 2-10 different designs at one time (Alonso-Camino et al., 2013; Duong et al., 2013). Future implementation of these high-throughput CAR generation methods will also require new high-throughput assays to screen larger numbers of CARs in ways that will be relevant to in vivo CAR activity. Such methods have the capability of further clarifying a set of optimal CAR design rules and lead to more rapid CAR development, especially for new antigen targets.

Most clinical trials and pre-clinical experiments to date have been performed using bulk and undefined T cell populations expressing the CAR which have been variable among patients (Brentjens et al., 2011). There is some focus now on better defining the optimal T cell subsets for use in therapy (Riddell et al., 2014). Pre-clinical studies have shown that there are subsets of T cells that lead to longer term CAR T cell persistence and the ability to control tumor growth. Combinations of CD4+ and CD8+ T cells together have been shown to be better than CD8+ T cells alone, and CD4+ and CD8+ naïve and central memory T cells (Tcm) appear to be more effective than effector memory T cells (Tem). Another general approach to augment CAR T cell function is to provide an accessory protein along with the CAR, either constitutively or inducibly-expressed upon T cell activation, creating so-called “armored CARs” (Morello et al., 2016; Pegram, Park, & Brentjens, 2014). Some of these accessory proteins include cytokines such as IL-12 or switch receptors that can bind to immunosuppressive proteins on tumor cells and lead to CAR T-cell co-stimulation (Liu et al., 2016). It is possible that certain accessory proteins will be optimal for patients with certain TMEs requiring a personalized CAR approach. Another common set of accessory genes is CAR suicide switches which allow for drug-mediated depletion of CAR T cells. These switches will be especially useful for Phase I trials testing the safety of new CARs with unknown toxicities and in the cases where the CARs target antigens found on normal cell populations that can be re-populated only after the CAR T cells are eliminated from the patient (Budde et al., 2013; Narayanan et al., 2011; X. Wang et al., 2011).

Finally, while CAR therapy is not yet FDA-approved, such approval is expected for CD19 CARs. There are currently over 50 registered CD19 CAR clinical trials across various institutions accounting for a treatment population of over 3300 patients. As CAR therapy is very complex involving genetic modification, culturing and expansion of a patient's cells, it will require major efforts for process standardization in specialized centers before bringing it to widespread use (Kaiser et al., 2015; X. Wang & Riviere, 2016).

4. Therapeutic cancer vaccines

The general purpose of a cancer vaccine is to elicit, expand or boost patients' tumor antigen-specific T cells and antibodies. In the therapeutic setting, vaccines are intended to activate the patient's immune cells that have already effectively lost the battle with cancer. Therapeutic cancer vaccines have been tested for many different cancers, in a variety of antigen and adjuvant combinations and delivery methods (Banday, Jeelani, & Hruby, 2015; Melero et al., 2014). They have targeted a plethora of different antigens including non-mutated shared tumor antigens as well as patient-specific mutated antigens. Many of these vaccines have elicited antigen-specific immune responses as defined by the production of antigen-specific T cells and antibodies in both pre-clinical and clinical settings. However, clinical outcomes have been largely disappointing. The major reason for this is now known to be the immune suppression established during the many years of tumor growth, which affects the ability of the vaccine to induce strong immune responses systemically and the ability of the induced immune response to function at the tumor site (Finn, 2003).

The first vaccine formulations were created from irradiated whole tumor cells, often also transfected to express immunostimulatory cytokines, that were expected to present shared antigens as well as patient-specific (unique) tumor antigens (Dranoff et al., 1993; Hanna & Peters, 1978). This approach had the advantage of not requiring knowledge of specific antigens expressed on the tumor and also of potentially eliciting a response to many different tumor antigens. Unfortunately these vaccines were largely unsuccessful as tumor cells likely had been previously immune-edited and/or may have even co-expressed immune suppressive molecules rendering them unlikely to elicit robust immune responses (Eager & Nemunaitis, 2005). There was some early clinical success using a related anti-idiotype vaccine approach for B-cell lymphomas in which antibodies generated from a patient's lymphoma cells were used as the vaccine antigen in combination with an adjuvant. Several patients were able to mount an immune response to their lymphomas and some even had complete disease regressions, however the vaccine ultimately did not meet clinical endpoints in a Phase III trial (Kwak et al., 1992; Levy et al., 2014).

As more and more tumor antigens were being identified, the field shifted to vaccines based on defined tumor antigens and one of a variety of immunostimulatory adjuvants, such as those that stimulate toll-like receptors (TLRs) or other pattern recognition receptors (PRRs) (Goutagny, Estornes, Hasan, Lebecque, & Caux, 2012; Parmiani et al., 2014; Steinhagen, Kinjo, Bode, & Klinman, 2011). The antigens were expected to be taken up by a patient's APCs, processed and presented to T cells. The soluble antigen was expected to stimulate antibody production by B cells. It became obvious that the patients' APC function was also compromised in the TME. This realization has led to new approaches, facilitated by the discovery of conditions under which human dendritic cells (DCs) could be generated and matured in vitro (Steinman & Banchereau, 2007). Patients' DCs were grown in vitro from peripheral blood monocytes, loaded with antigen, fully matured and re-infused into patients to stimulate anti-tumor immunity, primarily T cells (Nair, Archer, & Tedder, 2012; Tedder & Jansen, 2001). Other vaccine approaches have included DNA encoding the antigen to be taken up by DC in vivo, or loaded on DCs in vitro and presented to T cells (Rice, Ottensmeier, & Stevenson, 2008). Viruses, bacteria and yeast, engineered to express tumor antigens have also been used. Many of these approaches were capable of stimulating antigen-specific T cell and antibody responses and some even showed a marginal therapeutic benefit (Melero et al., 2014).

The first therapeutic cancer vaccine to be approved by the FDA, Sipleucel-T (trade-name Provenge) for prostate cancer combines in vitro generation and antigen (prostatic acid phosphatase, PAP) loading of patients' DCs with in vitro stimulation of patients' T cells. The whole mixture, antigen-loaded DC and in vitro stimulated T cells is then reinfused into the patient. FDA-approval was based on the overall survival benefits in 3 Phase II trials and a median 4.1 month survival increase in a Phase III clinical trial for patients with metastatic, asymptomatic hormone-refractory prostate cancer. There are ongoing clinical trials trying this vaccine in combination with various ICIs (Wei, Fong, & Small, 2015). While the positive clinical results with Provenge are notable, the company developing it, Dendreon, filed for bankruptcy soon after this FDA approval, highlighting the difficulty in bringing an expensive and patient-personalized therapy into widespread use.

For the future of therapeutic cancer vaccines active areas of investigation include the generation of new adjuvants, enhanced antigens and personalized peptide neoantigens (Banday et al., 2015; Gubin, Artyomov, Mardis, & Schreiber, 2015). Other promising approaches include the optimization of vaccine antigens to remove potential immune inhibitory epitopes and the use of multiple antigens to increase the breadth of the vaccine-elicited immune response (Cecil et al., 2014; Disis et al., 2013). Finally, the path forward for therapeutic cancer vaccines will depend on a more thorough understanding of the inhibitory signals in the TME and their application in combination with other immunomodulatory agents (van der Burg, Arens, Ossendorp, van Hall, & Melief, 2016).

5. Prophylactic cancer vaccines

The frontier in the field of cancer vaccines is the development of vaccines for the prevention of cancer (Finn & Beatty, 2016). The first FDA-approved prophylactic cancer vaccines prevent cancer indirectly by preventing infection with viruses known to cause cancer. Two of these vaccines, Gardasil and Cervarix, target and prevent human papilloma virus (HPV) infection, responsible for over 70% of cervical cancers, and the third, Recombivax HB, is for the prevention of hepatitis B infection, a major cause of liver cancer. The vaccines block infection largely through the induction of neutralizing antibodies. The results of several large clinical trials demonstrate their success at preventing cancer and the safety of this approach - 10 years for HPV and 25 years for hepatitis B (Schiller, Castellsague, & Garland, 2012; Trepo, 2014). As most cancers are not known to be caused by viruses there is great need to create vaccines to prevent non-viral cancers.

Currently, it is thought that ideal antigens for preventative cancer vaccines are those found on pre-malignant lesions and/or cancer stem cells (Kensler et al., 2016). Vaccinating individuals diagnosed with premalignant lesions who therefore are at an increased risk for cancer would strengthen immunosurveillance and lead to elimination of nascent tumors and their initiating cells at an earlier stage of tumor progression before a suppressive TME is likely to form. In addition to this requirement, it is also favorable to target antigens that are able to drive tumorigenesis, as cells that lose these driver genes would be less likely to survive.

In Table 3. we present a list of the subset of the top 75 tumor antigens from the NCI 2009 Workshop found to be expressed on premalignant lesions and/or stem cells, or are oncogenes and list the premalignant lesions that can be targeted. Many of these antigens have already been targeted by therapeutic vaccines. These antigens include the major families of the cancer/testis (CT) antigens including MAGE-A1, -A3 and –A4, NYESO-1 and GAGE which are found on early-stage invasive ductal carcinomas and ductal carcinoma in situ, squamous dysplasia leading to head and neck cancer, as well as esophageal squamous cell carcinoma in situ (Y. T. Chen, Panarelli, Piotti, & Yantiss, 2014; Piotti, Scognamiglio, Chiu, & Chen, 2013). Three other extensively studied tumor antigens found on various adenocarcinomas as well as on premalignant lesions are mesothelin, abnormally glycosylated Mucin 1 (MUC1) and hTERT. These antigens are expressed on pancreatic mucinous cysts and MUC1 and hTERT are also found in pancreatic intraepithelial neoplasms, precursors to pancreatic cancer (Adsay et al., 2002; Luttges, Feyerabend, Buchelt, Pacena, & Kloppel, 2002; Paini et al., 2014; S. Yonezawa, Higashi, Yamada, & Goto, 2008). Abnormally glycosylated MUC1 has also been found on breast ductal carcinomas in situ, as well as adenoma of the colon, a precursor to colon cancer (Ajioka, Watanabe, & Jass, 1997; Mommers et al., 1999). Recent studies to find new antigens expressed on cancer stem cells and premalignant lesions looking at colon adenoma and colorectal cancer microarrays identified 160 up-regulated genes compared to normal colon (Broussard et al., 2013). In another study researchers found that a multi-partite vaccine targeting three antigens on pre-invasive breast disease (Neu, IGFBP2, IGF-IR) could prevent breast cancer in a spontaneous mouse tumor model in which mice were treated after developing premalignant lesions (Disis et al., 2013). Other pre-clinical studies in mice with premalignant lesions have shown that common oncogene mutations that appear early and drive cancer formation could perhaps also be targeted by vaccines in animals including H-ras in carcinogen-induced tumors and EGFR in a lung cancer model (Ebben, Lubet, Gad, Disis, & You, 2016; Nasti et al., 2015). These common oncogenes in addition to others such as K-ras and p53 have been tested in therapeutic vaccines with limited efficacy but warrant further investigation as prophylactic vaccines (Carbone et al., 2005). Finally, another promising source of vaccine antigens are those targeted by spontaneous anti-tumor immune responses. Two such antigens, Cyclin B1 and SOX2, are found to be common targets of spontaneous immune responses for lung and prostate premalignant lesions and MGUS and MM, respectively (Dai et al., 2014; Kao et al., 2001; Spisek et al., 2007).

Table 3.

Therapeutic cancer vaccine antigens from the NCI 2009 workshop as candidates for prophylactic vaccines: expression in precancer.

NCI Ranka Antigen Oncogenica Cancer stem cell expressiona Premalignant lesion(s)b,c Cancer typec
1 WT1 yes yes nephrogenic rests Kidney
2 MUC1 yes yes PanIN, IPMN; BE; adenoma;bronchial preneoplasia; MGUS; AMM pancreatic, esophageal,colorectal, lung, multiple myeloma
3 LMP2 - yes ND -
4 HPV E6 E7 - some CIN, VIN cervical
5 EGFRvIII - yes ND -
6 HER-2/neu yes - DCIS; adenoma breast, colorectal
8 MAGE A3 - yes DCIS; SD; esophageal SCCIS breast, head and neck, esophageal
9 p53 non- mutant yes yes OL, OSMF; CIN; hyperplasia and dysplasia; schistosomal cystitis; SDIS; DH; dysplasia; DCIS oral, cervical, head and neck, bladder, lung, pancreatic, breast
10 NY-ESO-1 - yes DCIS; SD; SCIS breast, head and neck, esophageal
11 PSMA - - HGPIN prostate
13 CEA - - dysplasia, CIS; adenoma; IM cervical, colorectal, gastric
14 MelanA/MART1 - - BD, AK melanoma
15 Ras-mutant yes yes adenoma; EIN; BE; DH and dysplasia colorectal, endometrial, esophageal, pancreatic
17 p53 mutant yes some OL, OSMF; CIN; hyperplasia and dysplasia; schistosomal cystitis; SDIS; DH, dysplasia; DCIS; BE; AK, BD oral, cervical, head and neck, bladder, lung, pancreatic, breast, esophageal, melanoma
19 Bcr-abl yes yes ND -
20 Tyrosinase - - MIS, SSM melanoma
21 Survivin yes - OL; RIN; IPMT; BD oral, renal, pancreatic, melanoma
23 hTERT yes yes PanIN, IPMN; BD, AK pancreatic, melanoma
24 Sarcoma translocation breakpoints yes yes ND -
25 EphA2 yes - AAH, noninvasive bronchioloalveolar component of adenocarcinoma lung
27 ML-IAP yes - ND -
28 AFP - yes ND -
29 EpCAM - yes IM; IN gastric and esophageal
30 ERG (TMPRSS2 ETS fusion gene) yes - HGPIN prostate
32 PAX3 yes - neurofibroma nerve sheath, melanoma
33 ALK yes yes ND -
34 Androgen receptor yes - OL, OSMF; BPH, PIN oral, prostate
35 Cyclin B1 yes - bronchial preneoplasia; SD; DCIS lung, head and neck, breast
36 Polysialic Acid - - CIN; OL, OSMF cervical, oral
37 MYCN yes - ND -
38 RhoC yes - ND -
39 TRP-2 - yes ND -
42 Mesothelin - - PanIN, IPMN pancreatic
43 PSCA - - HGPIN prostate
44 MAGE A1 - yes DCIS; SD; SCCIS breast, head and neck, esophageal
45 sLe(a) - - adenoma; CIN colorectal, cervical
46 CYP1B1 - - PIN prostate
47 PLAC1 yes - DCIS breast
49 BORIS yes - ND -
50 Tn - - AK, BD; Paget's disease melanoma, breast
52 ETV6-AML yes - ND -
55 SART3 - - CIS gastric
56 STn - - adenoma, Crohn's colitis; IM colorectal, gastric
57 Carbonic anhydrase IX - - columnar cell metaplasia; BE, dysplasia colorectal, esophageal
58 PAX5 yes - ND -
59 OY-TES1 yes - ND -
66 B7H3 - - PIN prostate
a

Rankings from 2009 NCI Workshop reported in Cheever et al. 2009.

b

Premalignant lesions with validated expression on human tissue samples.

c

Listed cancer types correspond in order to listed premalignant lesions. Premalignant lesions of the same cancer tissue type are separated by commas while lesions of different tissue types are separated by semi-colons.

Patients at an increased risk for cancer stand to benefit most from prophylactic cancer vaccines. Statistics are available for many cancers indicating risk and timeframe for progression of premalignant lesions to cancer. Pre-clinical studies in mice engineered to develop spontaneous tumors show that such lesions either do not develop in vaccinated mice or do not progress to cancer (P. L. Beatty, Narayanan, Gariepy, Ranganathan, & Finn, 2010; Ebben et al., 2016). The increasing focus on early detection of cancer, including pre-malignant lesions, as well as identifying genetic and behavioral risk factors for cancer, can define candidate patient populations for prophylactic vaccination. A vaccine is a compelling alternative to current preventative measures, many of which rely on surgical and other invasive approaches (e.g. prophylactic mastectomies for patients with the BRCA1/2 mutation to prevent breast cancer and removal of CINs to prevent cervical or vulvar cancer) (S. Adams et al., 2011; Husemann et al., 2008; Singh et al., 2013). Indeed, when women diagnosed with premalignant vulvar neoplasias were given an HPV vaccine comprised of long peptides encoded by oncogenes E6 and E7 mixed with incomplete Freund's adjuvant, over 75% had significant responses with 47% clearing the lesions and maintaining a complete response for at least 24 months (de Vos van Steenwijk et al., 2012; de Vos van Steenwijk et al., 2014; Kenter et al., 2009). These responses correlated with the appearance of vaccine-induced HPV-specific CD4+ and CD8+ T cells. In 2013 our group undertook the first prophylactic cancer vaccine clinical trial targeting a non-viral antigen, MUC1, in patients with MUC1+ advanced adenomas of the colon (Kimura et al., 2013). The vaccine was well-tolerated with no adverse events occurring, and 47% of the patients produced high titers of anti-MUC1 antibodies and durable immune memory, an important requirement for a prophylactic vaccine. Of note the patients who did not respond already had high levels of circulating immunosuppressive myeloid derived suppressor cells (MDSCs), highlighting the great potential benefit to administering the vaccines even earlier. We are currently following up this trial with a larger scale, randomized Phase II trial for vaccine efficacy in preventing polyp recurrence (NCT02134925), and based on the documented safety we are seeking to carry out future trials even earlier in the pre-malignant process.

It would also be possible, albeit expensive, for patients with risk for cancer to receive a personalized prophylactic vaccine based on mutations discovered by sequencing of biopsies of pre-malignant lesions (Gubin et al., 2015). However, treating a patient prophylactically with a mutated antigen vaccine would require knowledge of the expression of antigen and its processing by APCs and presentation to T cells. Given the large number of shared tumor antigens for which this information is already known, it is much more practical for now to test the immunogenicity and safety of those tumor antigens as vaccines in the prophylactic setting (Finn, 2014).

6. Conclusions

In recent years, immunotherapy has been validated as an approach to treat cancer with many FDA-approved reagents becoming standard of care and current standard of care treatments being relegated to auxiliary roles. These successes have stimulated development of a multitude of new immunomodulatory drugs, drug combinations and advances in adoptive T cell therapy that promise to change the dire prognosis for the broad group of late-stage cancer patients. It is worth noting that several other promising immunotherapy approaches not covered in this review are being developed and are reviewed elsewhere including small molecule drugs capable of immune modulation and oncolytic viruses (J. L. Adams, Smothers, Srinivasan, & Hoos, 2015; Kaufman, Kohlhapp, & Zloza, 2015). The correct implementation of these therapies and determination of optimal therapeutic combinations will continue to be strengthened by basic research on the TME and the changing state of the immune system during tumor progression. Finally, a focused re-purposing of already developed therapeutic cancer vaccines to the prophylactic setting to treat patients at risk for cancer has the potential for the greatest overall impact by eliminating the cancer epidemic through immune prevention.

Abbreviations

AAH

atypical adenomatous hyperplasia

AK

actinic keratosis

AML

acute myelogenous leukemia

AMM

asymptomatic multiple myeloma

APC

antigen presenting cell

B-ALL

B cell acute lymphocytic leukemia

B-CLL

B cell chronic lymphocytic leukemia

BD

Bowen's disease

BE

Barrett's esophagus

B-NHL

B cell non-Hodgkin's lymphoma

BPH

benign prostatic hyperplasia

B-SLL

B cell small lymphocytic lymphoma

CAR

chimeric antigen receptor

CIN

cervical intraepithelial neoplasia

CIS

carcinoma in situ

CLL

chronic lymphocytic leukemia

CML

chronic myelogenous leukemia

DCIS

ductal carcinoma in situ

DH

ductal hyperplasia

EIN

endometrial intraepithelial neoplasia

HGPIN

high-grade prostatic intraepithelial neoplasia

HL

Hodgkin's lymphoma

HNSCC

head and neck squamous cell carcinoma

ICI

immune checkpoint inhibitor

IM

intestinal metaplasia

IN

intraepithelial neoplasia

IPMN

intraductal papillary mucinous neoplasm

mAb

monoclonal antibody

MGUS

monoclonal gammopathy of undetermined significance

MIS

melanoma in situ

MM

multiple myeloma

NA

not available

ND

not determined

NOS

not otherwise specified

NSCLC

non-small cell lung carcinoma

OL

oral leukoplakia

ORR

overall response rate

OSMF

oral submucous fibrosis

PanIN

pancreatic intraepithelial neoplasia

RCC

renal cell carcinoma

RIN

renal intratubular neoplasia

SD

squamous dysplasia

SSM

superficial spreading melanoma

TCR

T cell receptor

TME

tumor microenvironment

VIN

vulvar intraepithelial neoplasia

z

CD3-zeta fragment

Footnotes

Conflict of Interest Statement: The authors declare that there are no conflicts of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Adams JL, Smothers J, Srinivasan R, Hoos A. Big opportunities for small molecules in immuno-oncology. Nat Rev Drug Discov. 2015;14(9):603–622. doi: 10.1038/nrd4596. [DOI] [PubMed] [Google Scholar]
  2. Adams S, Greeder L, Reich E, Shao Y, Fosina D, Hanson N, et al. Jungbluth AA. Expression of cancer testis antigens in human BRCA-associated breast cancers: potential targets for immunoprevention? Cancer Immunol Immunother. 2011;60(7):999–1007. doi: 10.1007/s00262-011-1005-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Adsay V, Merati K, Andea A, Sarkar F, Hruban RH, Wilentz RE, et al. Klimstra DS. The dichotomy in the preinvasive neoplasia to invasive carcinoma sequence in the pancreas: differential expression of MUC1 and MUC2 supports the existence of two separate pathways of carcinogenesis. Mod Pathol. 2002;15(10):1087–1095. doi: 10.1097/01.MP.0000028647.98725.8B. [DOI] [PubMed] [Google Scholar]
  4. Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, et al. Sadelain M. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6(261):261ra151. doi: 10.1126/scitranslmed.3010162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ajioka Y, Watanabe H, Jass JR. MUC1 and MUC2 mucins in flat and polypoid colorectal adenomas. J Clin Pathol. 1997;50(5):417–421. doi: 10.1136/jcp.50.5.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Allard B, Pommey S, Smyth MJ, et al. Stagg J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res. 2013;19(20):5626–5635. doi: 10.1158/1078-0432.CCR-13-0545. [DOI] [PubMed] [Google Scholar]
  7. Alonso-Camino V, Sanchez-Martin D, Compte M, Nunez-Prado N, Diaz RM, Vile R, et al. Alvarez-Vallina L. CARbodies: Human Antibodies Against Cell Surface Tumor Antigens Selected From Repertoires Displayed on T Cell Chimeric Antigen Receptors. Mol Ther Nucleic Acids. 2013;2:e93. doi: 10.1038/mtna.2013.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Altvater B, Pscherer S, Juergens H, Rossig C. DAP10 Costimulates Chimeric Receptor-Mediated T Cell Responses to Tumor Antigen. ASH Annual Meeting Abstracts. 2005;106:3052. [Google Scholar]
  9. Antonioli L, Yegutkin GG, Pacher P, Blandizzi C, et al. Hasko G. Anti-CD73 in cancer immunotherapy: awakening new opportunities. Trends Cancer. 2016;2(2):95–109. doi: 10.1016/j.trecan.2016.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ascierto PA, et al. Marincola FM. 2015: The Year of Anti-PD-1/PD-L1s Against Melanoma and Beyond. EBioMedicine. 2015;2(2):92–93. doi: 10.1016/j.ebiom.2015.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Baixeras E, Huard B, Miossec C, Jitsukawa S, Martin M, Hercend T, et al. Piatier-Tonneau D. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J Exp Med. 1992;176(2):327–337. doi: 10.1084/jem.176.2.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Banday AH, Jeelani S, et al. Hruby VJ. Cancer vaccine adjuvants–recent clinical progress and future perspectives. Immunopharmacol Immunotoxicol. 2015;37(1):1–11. doi: 10.3109/08923973.2014.971963. [DOI] [PubMed] [Google Scholar]
  13. Barrett DM, Singh N, Porter DL, Grupp SA, et al. June CH. Chimeric antigen receptor therapy for cancer. Annu Rev Med. 2014;65:333–347. doi: 10.1146/annurev-med-060512-150254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bartkowiak T, Singh S, Yang G, Galvan G, Haria D, Ai M, et al. Curran MA. Unique potential of 4-1BB agonist antibody to promote durable regression of HPV+ tumors when combined with an E6/E7 eptide vaccine. Proc Natl Acad Sci U S A. 2015;112(38):E5290–5299. doi: 10.1073/pnas.1514418112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Beard RE, Zheng Z, Lagisetty KH, Burns WR, Tran E, Hewitt SM, et al. Morgan RA. Multiple chimeric antigen receptors successfully target chondroitin sulfate proteoglycan 4 in several different cancer histologies and cancer stem cells. J Immunother Cancer. 2014;2:25. doi: 10.1186/2051-1426-2-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. Vonderheide RH. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331(6024):1612–1616. doi: 10.1126/science.1198443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, et al. June CH. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res. 2014;2(2):112–120. doi: 10.1158/2326-6066.CIR-13-0170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Beatty PL, Narayanan S, Gariepy J, Ranganathan S, et al. Finn OJ. Vaccine against MUC1 antigen expressed in inflammatory bowel disease and cancer lessens colonic inflammation and prevents progression to colitis-associated colon cancer. Cancer Prev Res (Phila) 2010;3(4):438–446. doi: 10.1158/1940-6207.CAPR-09-0194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bennouna J, Sastre J, Arnold D, Osterlund P, Greil R, Van Cutsem E, et al. Investigators MLS. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol. 2013;14(1):29–37. doi: 10.1016/S1470-2045(12)70477-1. [DOI] [PubMed] [Google Scholar]
  20. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Wherry EJ. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37. doi: 10.1038/ni.1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Spigel DR. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med. 2015;373(2):123–135. doi: 10.1056/NEJMoa1504627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Wigginton JM. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–2465. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. Sadelain M. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra138. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J, et al. Sadelain M. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118(18):4817–4828. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Broussard EK, Kim R, Wiley JC, Marquez JP, Annis JE, Pritchard D, et al. Disis ML. Identification of putative immunologic targets for colon cancer prevention based on conserved gene upregulation from preinvasive to malignant lesions. Cancer Prev Res (Phila) 2013;6(7):666–674. doi: 10.1158/1940-6207.CAPR-12-0484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Budde LE, Berger C, Lin Y, Wang J, Lin X, Frayo SE, et al. Press OW. Combining a CD20 chimeric antigen receptor and an inducible caspase 9 suicide switch to improve the efficacy and safety of T cell adoptive immunotherapy for lymphoma. PLoS One. 2013;8(12):e82742. doi: 10.1371/journal.pone.0082742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Burdelya L, Kujawski M, Niu G, Zhong B, Wang T, Zhang S, et al. Yu H. Stat3 activity in melanoma cells affects migration of immune effector cells and nitric oxide-mediated antitumor effects. J Immunol. 2005;174(7):3925–3931. doi: 10.4049/jimmunol.174.7.3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Burnet M. Cancer; a biological approach. I. The processes of control. Br Med J. 1957;1(5022):779–786. doi: 10.1016/S0140-6736(57)91911-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, et al. Jakobsen BK. Identification of a Titin-derived HLA-A1-presented peptide as a crossreactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013;5(197):197ra103. doi: 10.1126/scitranslmed.3006034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Carbone DP, Ciernik IF, Kelley MJ, Smith MC, Nadaf S, Kavanaugh D, et al. Berzofsky JA. Immunization with mutant p53- and K-ras-derived peptides in cancer patients: immune response and clinical outcome. J Clin Oncol. 2005;23(22):5099–5107. doi: 10.1200/JCO.2005.03.158. [DOI] [PubMed] [Google Scholar]
  31. Cecil DL, Holt GE, Park KH, Gad E, Rastetter L, Childs J, et al. Disis ML. Elimination of IL-10-inducing T-helper epitopes from an IGFBP-2 vaccine ensures potent antitumor activity. Cancer Res. 2014;74(10):2710–2718. doi: 10.1158/0008-5472.CAN-13-3286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Chapman PB, D'Angelo SP, et al. Wolchok JD. Rapid eradication of a bulky melanoma mass with one dose of immunotherapy. N Engl J Med. 2015;372(21):2073–2074. doi: 10.1056/NEJMc1501894. [DOI] [PubMed] [Google Scholar]
  33. Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. Matrisian LM. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15(17):5323–5337. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Chen L, et al. Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13(4):227–242. doi: 10.1038/nri3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Chen TW, Razak AR, Bedard PL, Siu LL, et al. Hansen AR. A systematic review of immune-related adverse event reporting in clinical trials of immune checkpoint inhibitors. Ann Oncol. 2015;26(9):1824–1829. doi: 10.1093/annonc/mdv182. [DOI] [PubMed] [Google Scholar]
  36. Chen YT, Panarelli NC, Piotti KC, et al. Yantiss RK. Cancer-testis antigen expression in digestive tract carcinomas: frequent expression in esophageal squamous cell carcinoma and its precursor lesions. Cancer Immunol Res. 2014;2(5):480–486. doi: 10.1158/2326-6066.CIR-13-0124. [DOI] [PubMed] [Google Scholar]
  37. Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, Kim SC, et al. Wolchok JD. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS One. 2010;5(5):e10436. doi: 10.1371/journal.pone.0010436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Coley WB. The Treatment of Inoperable Sarcoma by Bacterial Toxins (the Mixed Toxins of the Streptococcus erysipelas and the Bacillus prodigiosus) Proc R Soc Med. 1910;3(Surg Sect):1–48. doi: 10.1001/jama.1898.92450090022001g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Condomines M, Arnason J, Benjamin R, Gunset G, Plotkin J, et al. Sadelain M. Tumor-Targeted Human T Cells Expressing CD28-Based Chimeric Antigen Receptors Circumvent CTLA-4 Inhibition. PLoS One. 2015;10(6):e0130518. doi: 10.1371/journal.pone.0130518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. 2013;342(6165):1432–1433. doi: 10.1126/science.342.6165.1432. [DOI] [PubMed] [Google Scholar]
  41. Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K, et al. Weinberg AD. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res. 2013;73(24):7189–7198. doi: 10.1158/0008-5472.CAN-12-4174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Dai L, Li J, Ortega R, Qian W, Casiano CA, et al. Zhang JY. Preferential autoimmune response in prostate cancer to cyclin B1 in a panel of tumor-associated antigens. J Immunol Res 2014. 2014:827827. doi: 10.1155/2014/827827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Brentjens R. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra225. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. de Vos van Steenwijk PJ, Ramwadhdoebe TH, Lowik MJ, van der Minne CE, Berends-van der Meer DM, Fathers LM, et al. van der Burg SH. A placebo-controlled randomized HPV16 synthetic long-peptide vaccination study in women with high-grade cervical squamous intraepithelial lesions. Cancer Immunol Immunother. 2012;61(9):1485–1492. doi: 10.1007/s00262-012-1292-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. de Vos van Steenwijk PJ, van Poelgeest MI, Ramwadhdoebe TH, Lowik MJ, Berends-van der Meer DM, van der Minne CE, et al. Kenter GG. The long-term immune response after HPV16 peptide vaccination in women with low-grade pre-malignant disorders of the uterine cervix: a placebo-controlled phase II study. Cancer Immunol Immunother. 2014;63(2):147–160. doi: 10.1007/s00262-013-1499-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, et al. Weichselbaum RR. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity. 2014;41(5):843–852. doi: 10.1016/j.immuni.2014.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn GP, Archambault JM, et al. Schreiber RD. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208(10):1989–2003. doi: 10.1084/jem.20101158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Disis ML, Gad E, Herendeen DR, Lai VP, Park KH, Cecil DL, et al. Lubet RA. A multiantigen vaccine targeting neu, IGFBP-2, and IGF-IR prevents tumor progression in mice with preinvasive breast disease. Cancer Prev Res (Phila) 2013;6(12):1273–1282. doi: 10.1158/1940-6207.CAPR-13-0182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Dranoff G, Jaffee E, Lazenby A, Golumbek P, Levitsky H, Brose K, et al. Mulligan RC. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci U S A. 1993;90(8):3539–3543. doi: 10.1073/pnas.90.8.3539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Dunn GP, Old LJ, et al. Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004a;21(2):137–148. doi: 10.1016/j.immuni.2004.07.017. [DOI] [PubMed] [Google Scholar]
  51. Dunn GP, Old LJ, et al. Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004b;22:329–360. doi: 10.1146/annurev.immunol.22.012703.104803. [DOI] [PubMed] [Google Scholar]
  52. Duong CP, Westwood JA, Yong CS, Murphy A, Devaud C, John LB, et al. Kershaw MH. Engineering T cell function using chimeric antigen receptors identified using a DNA library approach. PLoS One. 2013;8(5):e63037. doi: 10.1371/journal.pone.0063037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Eager R, et al. Nemunaitis J. GM-CSF gene-transduced tumor vaccines. Mol Ther. 2005;12(1):18–27. doi: 10.1016/j.ymthe.2005.02.012. [DOI] [PubMed] [Google Scholar]
  54. Ebben JD, Lubet RA, Gad E, Disis ML, et al. You M. Epidermal growth factor receptor derived peptide vaccination to prevent lung adenocarcinoma formation: An in vivo study in a murine model of EGFR mutant lung cancer. Mol Carcinog. 2016;55(11):1517–1525. doi: 10.1002/mc.22405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Eliopoulos AG, et al. Young LS. The role of the CD40 pathway in the pathogenesis and treatment of cancer. Curr Opin Pharmacol. 2004;4(4):360–367. doi: 10.1016/j.coph.2004.02.008. [DOI] [PubMed] [Google Scholar]
  56. Fan X, Quezada SA, Sepulveda MA, Sharma P, et al. Allison JP. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med. 2014;211(4):715–725. doi: 10.1084/jem.20130590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Fedorov VD, Themeli M, et al. Sadelain M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med. 2013;5(215):215ra172. doi: 10.1126/scitranslmed.3006597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Fielding AK, Richards SM, Chopra R, Lazarus HM, Litzow MR, Buck G, et al. Eastern Cooperative Oncology, G Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood. 2007;109(3):944–950. doi: 10.1182/blood-2006-05-018192. [DOI] [PubMed] [Google Scholar]
  59. Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3(8):630–641. doi: 10.1038/nri1150. [DOI] [PubMed] [Google Scholar]
  60. Finn OJ. Vaccines for cancer prevention: a practical and feasible approach to the cancer epidemic. Cancer Immunol Res. 2014;2(8):708–713. doi: 10.1158/2326-6066.CIR-14-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Finn OJ, et al. Beatty PL. Cancer immunoprevention. Curr Opin Immunol. 2016;39:52–58. doi: 10.1016/j.coi.2016.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, et al. Gajewski TF. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8 {alpha}+ dendritic cells. J Exp Med. 2011;208(10):2005–2016. doi: 10.1084/jem.20101159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Gajewski TF. The Next Hurdle in Cancer Immunotherapy: Overcoming the Non-T-Cell-Inflamed Tumor Microenvironment. Semin Oncol. 2015;42(4):663–671. doi: 10.1053/j.seminoncol.2015.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Galluzzi L, Buque A, Kepp O, Zitvogel L, et al. Kroemer G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell. 2015;28(6):690–714. doi: 10.1016/j.ccell.2015.10.012. [DOI] [PubMed] [Google Scholar]
  65. Garfall AL, Maus MV, Hwang WT, Lacey SF, Mahnke YD, Melenhorst JJ, et al. Stadtmauer EA. Chimeric Antigen Receptor T Cells against CD19 for Multiple Myeloma. N Engl J Med. 2015;373(11):1040–1047. doi: 10.1056/NEJMoa1504542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Goutagny N, Estornes Y, Hasan U, Lebecque S, et al. Caux C. Targeting pattern recognition receptors in cancer immunotherapy. Target Oncol. 2012;7(1):29–54. doi: 10.1007/s11523-012-0213-1. [DOI] [PubMed] [Google Scholar]
  67. Gross G, Waks T, et al. Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86(24):10024–10028. doi: 10.1073/pnas.86.24.10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Gubin MM, Artyomov MN, Mardis ER, et al. Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125(9):3413–3421. doi: 10.1172/JCI80008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Guedan S, Chen X, Madar A, Carpenito C, McGettigan SE, Frigault MJ, et al. June CH. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood. 2014;124(7):1070–1080. doi: 10.1182/blood-2013-10-535245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hamzah J, Nelson D, Moldenhauer G, Arnold B, Hammerling GJ, et al. Ganss R. Vascular targeting of anti-CD40 antibodies and IL-2 into autochthonous tumors enhances immunotherapy in mice. J Clin Invest. 2008;118(5):1691–1699. doi: 10.1172/JCI33201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Hanahan D, et al. Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  72. Hanna MG, Peters LC. Immunotherapy of established micrometastases with Bacillus Calmette-Guerin tumor cell vaccine. Cancer Res. 1978;38(1):204–209. [PubMed] [Google Scholar]
  73. Haso W, Lee DW, Shah NN, Stetler-Stevenson M, Yuan CM, Pastan IH, et al. Orentas RJ. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121(7):1165–1174. doi: 10.1182/blood-2012-06-438002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. He LZ, Prostak N, Thomas LJ, Vitale L, Weidlick J, Crocker A, et al. Keler T. Agonist anti-human CD27 monoclonal antibody induces T cell activation and tumor immunity in human CD27-transgenic mice. J Immunol. 2013;191(8):4174–4183. doi: 10.4049/jimmunol.1300409. [DOI] [PubMed] [Google Scholar]
  75. Hirschhorn-Cymerman D, Rizzuto GA, Merghoub T, Cohen AD, Avogadri F, Lesokhin AM, et al. Houghton AN. OX40 engagement and chemotherapy combination provides potent antitumor immunity with concomitant regulatory T cell apoptosis. J Exp Med. 2009;206(5):1103–1116. doi: 10.1084/jem.20082205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hombach AA, et al. Abken H. Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. Int J Cancer. 2011;129(12):2935–2944. doi: 10.1002/ijc.25960. [DOI] [PubMed] [Google Scholar]
  78. Horton HM, Bernett MJ, Peipp M, Pong E, Karki S, Chu SY, et al. Zhukovsky EA. Fc-engineered anti-CD40 antibody enhances multiple effector functions and exhibits potent in vitro and in vivo antitumor activity against hematologic malignancies. Blood. 2010;116(16):3004–3012. doi: 10.1182/blood-2010-01-265280. [DOI] [PubMed] [Google Scholar]
  79. Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, et al. Vignali DA. Role of LAG-3 in regulatory T cells. Immunity. 2004;21(4):503–513. doi: 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
  80. Huang YH, Zhu C, Kondo Y, Anderson AC, Gandhi A, Russell A, et al. Blumberg RS. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature. 2015;517(7534):386–390. doi: 10.1038/nature13848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, Rader C, et al. Riddell SR. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res. 2015;3(2):125–135. doi: 10.1158/2326-6066.CIR-14-0127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Husemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E, et al. Klein CA. Systemic spread is an early step in breast cancer. Cancer Cell. 2008;13(1):58–68. doi: 10.1016/j.ccr.2007.12.003. [DOI] [PubMed] [Google Scholar]
  83. Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I, et al. Kroczek RA. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature. 1999;397(6716):263–266. doi: 10.1038/16717. [DOI] [PubMed] [Google Scholar]
  84. Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, et al. Goldszmid RS. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–970. doi: 10.1126/science.1240527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Johnson LA, Scholler J, Ohkuri T, Kosaka A, Patel PR, McGettigan SE, et al. Maus MV. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med. 2015;7(275):275ra222. doi: 10.1126/scitranslmed.aaa4963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Kaiser AD, Assenmacher M, Schroder B, Meyer M, Orentas R, Bethke U, et al. Dropulic B. Towards a commercial process for the manufacture of genetically modified T cells for therapy. Cancer Gene Ther. 2015;22(2):72–78. doi: 10.1038/cgt.2014.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Kanamaru F, Youngnak P, Hashiguchi M, Nishioka T, Takahashi T, Sakaguchi S, et al. Azuma M. Costimulation via glucocorticoid-induced TNF receptor in both conventional and CD25+ regulatory CD4+ T cells. J Immunol. 2004;172(12):7306–7314. doi: 10.4049/jimmunol.172.12.7306. [DOI] [PubMed] [Google Scholar]
  88. Kao H, Marto JA, Hoffmann TK, Shabanowitz J, Finkelstein SD, Whiteside TL, et al. Finn OJ. Identification of cyclin B1 as a shared human epithelial tumor-associated antigen recognized by T cells. J Exp Med. 2001;194(9):1313–1323. doi: 10.1084/jem.194.9.1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Kaufman HL, Kohlhapp FJ, et al. Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015;14(9):642–662. doi: 10.1038/nrd4663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Kaufmann SH. Immunology's foundation: the 100-year anniversary of the Nobel Prize to Paul Ehrlich and Elie Metchnikoff. Nat Immunol. 2008;9(7):705–712. doi: 10.1038/ni0708-705. [DOI] [PubMed] [Google Scholar]
  91. Kawalekar OU, O'Connor RS, Fraietta JA, Guo L, McGettigan SE, Posey AD, Jr, et al. June CH. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. Immunity. 2016;44(2):380–390. doi: 10.1016/j.immuni.2016.01.021. [DOI] [PubMed] [Google Scholar]
  92. Kebriaei P, Singh H, Huls MH, Figliola MJ, Bassett R, Olivares S, et al. Cooper LJ. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J Clin Invest. 2016;126(9):3363–3376. doi: 10.1172/JCI86721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kensler TW, Spira A, Garber JE, Szabo E, Lee JJ, Dong Z, et al. Lippman SM. Transforming Cancer Prevention through Precision Medicine and Immune-oncology. Cancer Prev Res (Phila) 2016;9(1):2–10. doi: 10.1158/1940-6207.CAPR-15-0406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, et al. Melief CJ. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361(19):1838–1847. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]
  95. Khalil DN, Smith EL, Brentjens RJ, et al. Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13(5):273–290. doi: 10.1038/nrclinonc.2016.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Kim JE, Patel MA, Mangraviti A, Kim ES, Theodros D, Velarde E, et al. Lim M. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin Cancer Res. 2016 doi: 10.1158/1078-0432.CCR-15-1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Kimura T, McKolanis JR, Dzubinski LA, Islam K, Potter DM, Salazar AM, et al. Finn OJ. MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study. Cancer Prev Res (Phila) 2013;6(1):18–26. doi: 10.1158/1940-6207.CAPR-12-0275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Kloss CC, Condomines M, Cartellieri M, Bachmann M, et al. Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2013;31(1):71–75. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, Yamaguchi T, et al. Sakaguchi S. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med. 2005;202(7):885–891. doi: 10.1084/jem.20050940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Kocak E, Lute K, Chang X, May KF, Jr, Exten KR, Zhang H, et al. Liu Y. Combination therapy with anti-CTL antigen-4 and anti-4-1BB antibodies enhances cancer immunity and reduces autoimmunity. Cancer Res. 2006;66(14):7276–7284. doi: 10.1158/0008-5472.CAN-05-2128. [DOI] [PubMed] [Google Scholar]
  101. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, et al. Rosenberg SA. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33(6):540–549. doi: 10.1200/JCO.2014.56.2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Kohler G, et al. Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256(5517):495–497. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
  103. Kohrt HE, Thielens A, Marabelle A, Sagiv-Barfi I, Sola C, Chanuc F, et al. Andre P. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood. 2014;123(5):678–686. doi: 10.1182/blood-2013-08-519199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Krummel MF, et al. Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995;182(2):459–465. doi: 10.1210/me.2014-1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Kwak LW, Campbell MJ, Czerwinski DK, Hart S, Miller RA, et al. Levy R. nduction of immune responses in patients with B-cell lymphoma against the surface-immunoglobulin idiotype expressed by their tumors. N Engl J Med. 1992;327(17):1209–1215. doi: 10.1056/NEJM199210223271705. [DOI] [PubMed] [Google Scholar]
  106. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373(1):23–34. doi: 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Le Mercier I, Chen W, Lines JL, Day M, Li J, Sergent P, et al. Wang L. VISTA Regulates the Development of Protective Antitumor Immunity. Cancer Res. 2014;74(7):1933–1944. doi: 10.1158/0008-5472.CAN-13-1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Leach DR, Krummel MF, et al. Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734–1736. doi: 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
  109. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. Mackall CL. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–195. doi: 10.1182/blood-2014-05-552729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. Mackall CL. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–528. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Lee HW, Park SJ, Choi BK, Kim HH, Nam KO, et al. Kwon BS. 4-1BB promotes the survival of CD8+ T lymphocytes by increasing expression of Bcl-xL and Bfl-1. J Immunol. 2002;169(9):4882–4888. doi: 10.4049/jimmunol.169.9.4882. [DOI] [PubMed] [Google Scholar]
  112. Levy R, Ganjoo KN, Leonard JP, Vose JM, Flinn IW, Ambinder RF, et al. Denney DW., Jr Active idiotypic vaccination versus control immunotherapy for follicular lymphoma. J Clin Oncol. 2014;32(17):1797–1803. doi: 10.1200/JCO.2012.43.9273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Linch SN, McNamara MJ, et al. Redmond WL. OX40 Agonists and Combination Immunotherapy: Putting the Pedal to the Metal. Front Oncol. 2015;5:34. doi: 10.3389/fonc.2015.00034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Lines JL, Pantazi E, Mak J, Sempere LF, Wang L, O'Connell S, et al. Noelle R. VISTA is an immune checkpoint molecule for human T cells. Cancer Res. 2014;74(7):1924–1932. doi: 10.1158/0008-5472.CAN-13-1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC, et al. Zhao Y. Affinity-Tuned ErbB2 or EGFR Chimeric Antigen Receptor T Cells Exhibit an Increased Therapeutic Index against Tumors in Mice. Cancer Res. 2015;75(17):3596–3607. doi: 10.1158/0008-5472.CAN-15-0159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Liu X, Ranganathan R, Jiang S, Fang C, Sun J, Kim S, et al. Moon EK. A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors. Cancer Res. 2016;76(6):1578–1590. doi: 10.1158/0008-5472.CAN-15-2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Lohmueller JJ, Sato S, Popova L, Chu IM, Tucker MA, Barberena R, et al. Finn OJ. Antibodies elicited by the first non-viral prophylactic cancer vaccine show tumor-specificity and immunotherapeutic potential. Sci Rep. 2016;6:31740. doi: 10.1038/srep31740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, et al. Sotiriou C. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol. 2014;25(8):1544–1550. doi: 10.1093/annonc/mdu112. [DOI] [PubMed] [Google Scholar]
  119. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, et al. Mackall CL. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015;21(6):581–590. doi: 10.1038/nm.3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Luheshi NM, Coates-Ulrichsen J, Harper J, Mullins S, Sulikowski MG, Martin P, et al. Wilkinson RW. Transformation of the tumour microenvironment by a CD40 agonist antibody correlates with improved responses to PD-L1 blockade in a mouse orthotopic pancreatic tumour model. Oncotarget. 2016;7(14):18508–18520. doi: 10.18632/oncotarget.7610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Luttges J, Feyerabend B, Buchelt T, Pacena M, et al. Kloppel G. The mucin profile of noninvasive and invasive mucinous cystic neoplasms of the pancreas. Am J Surg Pathol. 2002;26(4):466–471. doi: 10.1097/00000478-200204000-00008. [DOI] [PubMed] [Google Scholar]
  122. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. North-East Japan Study G. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362(25):2380–2388. doi: 10.1056/NEJMoa0909530. [DOI] [PubMed] [Google Scholar]
  123. Maher J, Brentjens RJ, Gunset G, Riviere I, et al. Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor. Nat Biotechnol. 2002;20(1):70–75. doi: 10.1038/nbt0102-70. [DOI] [PubMed] [Google Scholar]
  124. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Grupp SA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. May KF, Jr, Chen L, Zheng P, Liu Y. Anti-4-1BB monoclonal antibody enhances rejection of large tumor burden by promoting survival but not clonal expansion of tumor-specific CD8+ T cells. Cancer Res. 2002;62(12):3459–3465. [PubMed] [Google Scholar]
  126. Melero I, Gaudernack G, Gerritsen W, Huber C, Parmiani G, Scholl S, et al. Mellstedt H. Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol. 2014;11(9):509–524. doi: 10.1038/nrclinonc.2014.111. [DOI] [PubMed] [Google Scholar]
  127. Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE, et al. Chen L. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med. 1997;3(6):682–685. doi: 10.1038/nm0697-682. [DOI] [PubMed] [Google Scholar]
  128. Michot JM, Bigenwald C, Champiat S, Collins M, Carbonnel F, Postel-Vinay S, et al. Lambotte O. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer. 2016;54:139–148. doi: 10.1016/j.ejca.2015.11.016. [DOI] [PubMed] [Google Scholar]
  129. Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26(305):234–241. doi: 10.1259/0007-1285-26-305-234. [DOI] [PubMed] [Google Scholar]
  130. Mommers EC, Leonhart AM, von Mensdorff-Pouilly S, Schol DJ, Hilgers J, Meijer CJ, et al. van Diest PJ. Aberrant expression of MUC1 mucin in ductal hyperplasia and ductal carcinoma In situ of the breast. Int J Cancer. 1999;84(5):466–469. doi: 10.1002/(sici)1097-0215(19991022)84:5<466::aid-ijc3>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
  131. Morello A, Sadelain M, et al. Adusumilli PS. Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov. 2016;6(2):133–146. doi: 10.1158/2159-8290.CD-15-0583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, et al. Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–851. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. CheckMate I. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med. 2015;373(19):1803–1813. doi: 10.1056/NEJMoa1510665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Nair S, Archer GE, et al. Tedder TF. Isolation and generation of human dendritic cells. Curr Protoc Immunol. 2012;Chapter 7 doi: 10.1002/0471142735.im0732s99. Unit7 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Narayanan P, Lapteva N, Seethammagari M, Levitt JM, Slawin KM, et al. Spencer DM. A composite MyD88/CD40 switch synergistically activates mouse and human dendritic cells for enhanced antitumor efficacy. J Clin Invest. 2011;121(4):1524–1534. doi: 10.1172/JCI44327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Nasti TH, Rudemiller KJ, Cochran JB, Kim HK, Tsuruta Y, Fineberg NS, et al. Timares L. Immunoprevention of chemical carcinogenesis through early recognition of oncogene mutations. J Immunol. 2015;194(6):2683–2695. doi: 10.4049/jimmunol.1402125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Nghiem PT, Bhatia S, Lipson EJ, Kudchadkar RR, Miller NJ, Annamalai L, et al. Cheever MA. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N Engl J Med. 2016;374(26):2542–2552. doi: 10.1056/NEJMoa1603702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Nocentini G, et al. Riccardi C. GITR: a multifaceted regulator of immunity belonging to the tumor necrosis factor receptor superfamily. Eur J Immunol. 2005;35(4):1016–1022. doi: 10.1002/eji.200425818. [DOI] [PubMed] [Google Scholar]
  139. Oh SS, Moon C, Kim DH, Song H, Park S, Fu Y, et al. Kim KD. Adenovirally delivered IFN-beta exerts antitumor effects through transient T-lymphocyte depletion and Ag-specific T-cell proliferation. Int J Mol Med. 2012;29(6):1153–1157. doi: 10.3892/ijmm.2012.936. [DOI] [PubMed] [Google Scholar]
  140. Ohigashi Y, Sho M, Yamada Y, Tsurui Y, Hamada K, Ikeda N, et al. Nakajima Y. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin Cancer Res. 2005;11(8):2947–2953. doi: 10.1158/1078-0432.CCR-04-1469. [DOI] [PubMed] [Google Scholar]
  141. Okudaira K, Hokari R, Tsuzuki Y, Okada Y, Komoto S, Watanabe C, et al. Miura S. Blockade of B7-H1 or B7-DC induces an anti-tumor effect in a mouse pancreatic cancer model. Int J Oncol. 2009;35(4):741–749. doi: 10.3892/ijo-00000387. [DOI] [PubMed] [Google Scholar]
  142. Paini M, Crippa S, Partelli S, Scopelliti F, Tamburrino D, Baldoni A, et al. Falconi M. Molecular pathology of intraductal papillary mucinous neoplasms of the pancreas. World J Gastroenterol. 2014;20(29):10008–10023. doi: 10.3748/wjg.v20.i29.10008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Pan PY, Zang Y, Weber K, Meseck ML, et al. Chen SH. OX40 ligation enhances primary and memory cytotoxic T lymphocyte responses in an immunotherapy for hepatic colon metastases. Mol Ther. 2002;6(4):528–536. doi: 10.1006/mthe.2002.0699. [DOI] [PubMed] [Google Scholar]
  144. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Park JH, Riviere I, Wang X, Bernal YJ, Yoo S, Purdon T, et al. Brentjens RJ. CD19-Targeted 19-28z CAR Modified Autologous T Cells Induce High Rates of Complete Remission and Durable Responses in Adult Patients with Relapsed, Refractory B-Cell ALL. Blood. 2014;124:382. [Google Scholar]
  146. Park JJ, Omiya R, Matsumura Y, Sakoda Y, Kuramasu A, Augustine MM, et al. Tamada K. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood. 2010;116(8):1291–1298. doi: 10.1182/blood-2010-01-265975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, et al. Maio M. Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother. 2014;10(11):3175–3178. doi: 10.4161/hv.29418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Pedicord VA, Montalvo W, Leiner IM, et al. Allison JP. Single dose of anti-CTLA-4 enhances CD8+ T-cell memory formation, function, and maintenance. Proc Natl Acad Sci U S A. 2011;108(1):266–271. doi: 10.1073/pnas.1016791108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Pegram HJ, Park JH, et al. Brentjens RJ. CD28z CARs and armored CARs. Cancer J. 2014;20(2):127–133. doi: 10.1097/PPO.0000000000000034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Piotti KC, Scognamiglio T, Chiu R, et al. Chen YT. Expression of cancer/testis (CT) antigens in squamous cell carcinoma of the head and neck: evaluation as markers of squamous dysplasia. Pathol Res Pract. 2013;209(11):721–726. doi: 10.1016/j.prp.2013.08.004. [DOI] [PubMed] [Google Scholar]
  151. Porter DL, Levine BL, Kalos M, Bagg A, et al. June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Wolchok JD. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–931. doi: 10.1056/NEJMoa1112824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al. Hodi FS. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–2017. doi: 10.1056/NEJMoa1414428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, et al. Vogelzang NJ. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515(7528):558–562. doi: 10.1038/nature13904. [DOI] [PubMed] [Google Scholar]
  155. Redman JM, Hill EM, AlDeghaither D, et al. Weiner LM. Mechanisms of action of therapeutic antibodies for cancer. Mol Immunol. 2015;67(2 Pt A):28–45. doi: 10.1016/j.molimm.2015.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Rice J, Ottensmeier CH, et al. Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev Cancer. 2008;8(2):108–120. doi: 10.1038/nrc2326. [DOI] [PubMed] [Google Scholar]
  157. Riddell SR, Sommermeyer D, Berger C, Liu LS, Balakrishnan A, Salter A, et al. Turtle CJ. Adoptive therapy with chimeric antigen receptor-modified T cells of defined subset composition. Cancer J. 2014;20(2):141–144. doi: 10.1097/PPO.0000000000000036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Chan TA. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128. doi: 10.1126/science.aaa1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. investigators, K Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372(26):2521–2532. doi: 10.1056/NEJMoa1503093. [DOI] [PubMed] [Google Scholar]
  160. Robert L, Tsoi J, Wang X, Emerson R, Homet B, Chodon T, et al. Ribas A. CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res. 2014;20(9):2424–2432. doi: 10.1158/1078-0432.CCR-13-2648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Rogers PR, Song J, Gramaglia I, Killeen N, et al. Croft M. OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity. 2001;15(3)(01):445–455. 00191–1. doi: 10.1016/S1074-7613. [DOI] [PubMed] [Google Scholar]
  162. Romagne F, Andre P, Spee P, Zahn S, Anfossi N, Gauthier L, et al. Wagtmann N. Preclinical characterization of 1-7F9, a novel human anti-KIR receptor therapeutic antibody that augments natural killer-mediated killing of tumor cells. Blood. 2009;114(13):2667–2677. doi: 10.1182/blood-2009-02-206532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Ronchetti S, Nocentini G, Bianchini R, Krausz LT, Migliorati G, et al. Riccardi C. Glucocorticoid-induced TNFR-related protein lowers the threshold of CD28 costimulation in CD8+ T cells. J Immunol. 2007;179(9):5916–5926. doi: 10.4049/jimmunol.179.9.5916. [DOI] [PubMed] [Google Scholar]
  164. Roybal KT, Rupp LJ, Morsut L, Walker WJ, McNally KA, Park JS, et al. Lim WA. Precision Tumor Recognition by T Cells With Combinatorial Antigen-Sensing Circuits. Cell. 2016;164(4):770–779. doi: 10.1016/j.cell.2016.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Sabatos CA, Chakravarti S, Cha E, Schubart A, Sanchez-Fueyo A, Zheng XX, et al. Kuchroo VK. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol. 2003;4(11):1102–1110. doi: 10.1038/ni988. [DOI] [PubMed] [Google Scholar]
  166. Sadelain M, Brentjens R, et al. Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3(4):388–398. doi: 10.1158/2159-8290.CD-12-0548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, et al. Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207(10):2187–2194. doi: 10.1084/jem.20100643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, et al. Dotti G. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121(5):1822–1826. doi: 10.1172/JCI46110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Wolchok JD. Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J Clin Oncol. 2015;33(17):1889–1894. doi: 10.1200/JCO.2014.56.2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Schaer DA, Budhu S, Liu C, Bryson C, Malandro N, Cohen A, et al. Wolchok JD. GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res. 2013;1(5):320–331. doi: 10.1158/2326-6066.CIR-13-0086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Schiller JT, Castellsague X, et al. Garland SM. A review of clinical trials of human papillomavirus prophylactic vaccines. Vaccine. 2012;30(5):F123–138. doi: 10.1016/j.vaccine.2012.04.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Schuster SJ, Svoboda J, Dwivedy Nasta S, Porter DL, Chong EA, Mahnke Y, et al. June CH. Phase IIa Trial of Chimeric Antigen Receptor Modified T Cells Directed Against CD19 (CTL019) in Patients with Relapsed or Refractory CD19+ Lymphomas. Blood. 2014;124:3087. [Google Scholar]
  173. Scott AM, Wolchok JD, et al. Old LJ. Antibody therapy of cancer. Nat Rev Cancer. 2012;12(4):278–287. doi: 10.1038/nrc3236. [DOI] [PubMed] [Google Scholar]
  174. Shaw AT, Kim DW, Nakagawa K, Seto T, Crino L, Ahn MJ, et al. Janne PA. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368(25):2385–2394. doi: 10.1056/NEJMoa1214886. [DOI] [PubMed] [Google Scholar]
  175. Shuford WW, Klussman K, Tritchler DD, Loo DT, Chalupny J, Siadak AW, et al. Mittler RS. 4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses. J Exp Med. 1997;186(1):47–55. doi: 10.1084/jem.186.1.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Singh K, Lester J, Karlan B, Bresee C, Geva T, et al. Gordon O. Impact of family history on choosing risk-reducing surgery among BRCA mutation carriers. Am J Obstet Gynecol. 2013;208(4):329 e321–326. doi: 10.1016/j.ajog.2013.01.026. [DOI] [PubMed] [Google Scholar]
  177. Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Gajewski TF. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–1089. doi: 10.1126/science.aac4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Chan TA. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–2199. doi: 10.1056/NEJMoa1406498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Spisek R, Kukreja A, Chen LC, Matthews P, Mazumder A, Vesole D, et al. Dhodapkar MV. Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy. J Exp Med. 2007;204(4):831–840. doi: 10.1084/jem.20062387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Spranger S, Bao R, et al. Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–235. doi: 10.1038/nature14404. [DOI] [PubMed] [Google Scholar]
  181. Starck L, Scholz C, Dorken B, et al. Daniel PT. Costimulation by CD137/4-1BB inhibits T cell apoptosis and induces Bcl-xL and c-FLIP(short) via phosphatidylinositol 3-kinase and AKT/protein kinase B. Eur J Immunol. 2005;35(4):1257–1266. doi: 10.1002/eji.200425686. [DOI] [PubMed] [Google Scholar]
  182. Steinhagen F, Kinjo T, Bode C, et al. Klinman DM. TLR-based immune adjuvants. Vaccine. 2011;29(17):3341–3355. doi: 10.1016/j.vaccine.2010.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449(7161):419–426. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
  184. Tedder TF, et al. Jansen PJ. Isolation and generation of human dendritic cells. Curr Protoc Immunol. 2001;Chapter 7(Unit 7):32. doi: 10.1002/0471142735.im0732s23. [DOI] [PubMed] [Google Scholar]
  185. Thomas LJ, He LZ, Marsh H, et al. Keler T. Targeting human CD27 with an agonist antibody stimulates T-cell activation and antitumor immunity. Oncoimmunology. 2014;3(1):e27255. doi: 10.4161/onci.27255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG, et al. Press OW. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood. 2012;119(17):3940–3950. doi: 10.1182/blood-2011-10-387969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Sznol M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Topalian SL, Taube JM, Anders RA, et al. Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275–287. doi: 10.1038/nrc.2016.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Trepo C. A brief history of hepatitis milestones. Liver Int. 2014;34(1):29–37. doi: 10.1111/liv.12409. [DOI] [PubMed] [Google Scholar]
  190. Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al. Garraway LA. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350(6257):207–211. doi: 10.1126/science.aad0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. van der Burg SH, Arens R, Ossendorp F, van Hall T, et al. Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer. 2016;16(4):219–233. doi: 10.1038/nrc.2016.16. [DOI] [PubMed] [Google Scholar]
  192. Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, et al. Zitvogel L. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi: 10.1126/science.aad1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Waitz R, Fasso M, et al. Allison JP. CTLA-4 blockade synergizes with cryoablation to mediate tumor rejection. Oncoimmunology. 2012;1(4):544–546. doi: 10.4161/onci.19442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, et al. Bluestone JA. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1(5):405–413. doi: 10.1016/1074-7613(94)90071-X. [DOI] [PubMed] [Google Scholar]
  195. Wang L, Rubinstein R, Lines JL, Wasiuk A, Ahonen C, Guo Y, et al. Noelle RJ. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208(3):577–592. doi: 10.1084/jem.20100619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Wang LC, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, et al. Albelda SM. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res. 2014;2(2):154–166. doi: 10.1158/2326-6066.CIR-13-0027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Wang X, Chang WC, Wong CW, Colcher D, Sherman M, Ostberg JR, et al. Jensen MC. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood. 2011;118(5):1255–1263. doi: 10.1182/blood-2011-02-337360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Wang X, et al. Riviere I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics. 2016;3:16015. doi: 10.1038/mto.2016.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, et al. Mak TW. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270(5238):985–988. doi: 10.1126/science.270.5238.985. [DOI] [PubMed] [Google Scholar]
  200. Wei XX, Fong L, et al. Small EJ. Prostate Cancer Immunotherapy with Sipuleucel-T: Current Standards and Future Directions. Expert Rev Vaccines. 2015;14(12):1529–1541. doi: 10.1586/14760584.2015.1099437. [DOI] [PubMed] [Google Scholar]
  201. White AL, Chan HT, Roghanian A, French RR, Mockridge CI, Tutt AL, et al. Glennie MJ. Interaction with FcgammaRIIB is critical for the agonistic activity of anti-CD40 monoclonal antibody. J Immunol. 2011;187(4):1754–1763. doi: 10.4049/jimmunol.1101135. [DOI] [PubMed] [Google Scholar]
  202. Wilkie S, Picco G, Foster J, Davies DM, Julien S, Cooper L, et al. Maher J. Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J Immunol. 2008;180(7):4901–4909. doi: 10.4049/jimmunol.180.7.4901. [DOI] [PubMed] [Google Scholar]
  203. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, et al. Sakaguchi S. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322(5899):271–275. doi: 10.1126/science.1160062. [DOI] [PubMed] [Google Scholar]
  204. Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al. Vignali DA. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–927. doi: 10.1158/0008-5472.CAN-11-1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Yonezawa A, Dutt S, Chester C, Kim J, et al. Kohrt HE. Boosting Cancer Immunotherapy with Anti-CD137 Antibody Therapy. Clin Cancer Res. 2015;21(14):3113–3120. doi: 10.1158/1078-0432.CCR-15-0263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Yonezawa S, Higashi M, Yamada N, et al. Goto M. Precursor lesions of pancreatic cancer. Gut Liver. 2008;2(3):137–154. doi: 10.5009/gnl.2008.2.3.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Zamarin D, Holmgaard RB, Subudhi SK, Park JS, Mansour M, Palese P, et al. Allison JP. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci Transl Med. 2014;6(226):226ra232. doi: 10.1126/scitranslmed.3008095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Zhong XS, Matsushita M, Plotkin J, Riviere I, et al. Sadelain M. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther. 2010;18(2):413–420. doi: 10.1038/mt.2009.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, et al. Kuchroo VK. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6(12):1245–1252. doi: 10.1038/ni1271. [DOI] [PubMed] [Google Scholar]

RESOURCES