Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 May 1.
Published in final edited form as: Immunotherapy. 2014 Jul;6(7):833–852. doi: 10.2217/imt.14.51

Targeting CD8+ T-cell tolerance for cancer immunotherapy

Stephanie R Jackson 1,2, Jinyun Yuan 1,2, Ryan M Teague 1,2,*
PMCID: PMC4215555  NIHMSID: NIHMS635914  PMID: 25290416

Abstract

In the final issue of Science in 2013, the American Association of Science recognized progress in the field of cancer immunotherapy as the ‘Breakthrough of the Year.’ The achievements were actually twofold, owing to the early success of genetically engineered chimeric antigen receptors (CAR) and to the mounting clinical triumphs achieved with checkpoint blockade antibodies. While fundamentally very different, the common thread of these independent strategies is the ability to prevent or overcome mechanisms of CD8+ T-cell tolerance for improved tumor immunity. Here we discuss how circumventing T-cell tolerance has provided experimental insights that have guided the field of clinical cancer immunotherapy to a place where real breakthroughs can finally be claimed.

Keywords: adoptive cell transfer, cancer immunotherapy, cancer vaccine, CD8+ T-cell checkpoint blockade, chimeric antigen receptor, lymphodepletion, tolerance

Background

The goal of cancer immunotherapy is to enhance antitumor responses by a patient’s own immune system. Since the earliest days of immunotherapy, it has been appreciated that immunostimulation is required to elicit endogenous immunity against cancer. This began in the 1890s with Coley’s recognition of an apparent relationship between infection and cancer regression [1], which led to decades of effort aimed at developing cancer vaccines with nonspecific immune adjuvants. While this general approach was largely unsuccessful, it was instrumental to the recognition that specific responses would be required for therapy. Evidence for the role of cellular immunity was later demonstrated in 1955 [2], ultimately leading to early efforts at cancer immunotherapy by adoptive transfer of lymphocytes from immunized donors and extensive energy directed toward characterizing lymphocyte subpopulations with antitumor reactivity. Despite contributions by other immune cell subsets, CD8+ T cells have emerged as the predominant effector in most cancer immunotherapy settings. Thus, many immunotherapeutic strategies are dedicated to stimulating, enhancing and maintaining responses by tumor-reactive CD8+ T-cells.

A major obstacle to eliciting antitumor CD8+ T-cell immunity is tolerance. Immune tolerance exists to protect healthy tissues from immune-mediated attack. Because tumors commonly express self-antigens characteristic of the healthy tissue from which they are derived, mechanisms of tolerance often prevent appropriate immune responses toward malignant cells. CD8+ T-cell tolerance is broadly categorized into central and peripheral tolerance. In central tolerance, T cells specific for self-antigen are deleted in the thymus before they gain access to peripheral tissues. In contrast, peripheral tolerance serves to attenuate responses by T cells specific for self-antigens not expressed in the thymus, or expressed at such low levels that self-reactive T cells escape negative selection. Since central tolerance effectively creates holes in the tumor-reactive T-cell repertoire by deletion of self-reactive cells, the majority of immunotherapeutic approaches are targeted at preventing or overcoming peripheral tolerance. The mechanisms of peripheral T-cell tolerance are diverse, and can involve the specific deletion of T cells upon engagement of antigen expressed in the periphery by healthy tissues or malignant cells [3]. Alternatively, induction of T-cell dysfunction or anergy can be achieved by an array of stimuli emanating from inhibitory ligands, lack of costimulation, enzymes or the influences of regulatory cell populations [4]. While these processes protect healthy tissues from T-cell-mediated destruction, they represent major obstacles for eliciting immune responses against cancer. Thus, the overarching goal of immunotherapy is to avoid or disable this plethora of tolerizing forces to unleash the power of CD8+ T cells against cancer, while taking care to minimize and manage the inevitable autoimmunity that can arise from such endeavors. Here, we explore the myriad efforts to overcome CD8+ T-cell tolerance to provide durable immunotherapy in patients with cancer, and discuss how past lessons are informing current clinical successes.

Cytokines

Much of the early enthusiasm for cancer immunotherapy arose from the promising results of cytokine-based immunotherapy. The principle of cytokine immunotherapy for cancer is that these small proteins can coordinately regulate the growth and differentiation of lymphocytes, creating a host environment that promotes tumor regression. We have focused on the common gamma chain cytokines, which have well-defined roles in sustaining CD8+ T cells numbers and function. Here we briefly review efforts to harness these cytokines clinically, and highlight the technological advances that exploit their beneficial properties while avoiding the adverse effects that have limited similar application in the past.

IL-2

Initially called T-cell growth factor, IL-2 was first identified as a product of activated T cells that enhanced the proliferation and function of other T cells [5]. These attributes provided the original rationale for exploring IL-2 as an agent for cancer immunotherapy [68]. Indeed, high dose IL-2 was found to have clinical efficacy in some patients and was ultimately heralded as the first real success for immunotherapy [9]. However, success with IL-2 as a monotherapy was largely limited to metastatic renal cancer and melanoma, and achieved complete remissions in only a minority (5–10%) of patients, with durable responses in 70% of those patients that experienced complete regression [10].

Since its original characterization, roles for IL-2 in promoting CD8+ T-cell expansion, survival, memory cell development, cytolytic activity and even rescue of CD8+ T-cell tolerance have all been reported [11,12]. Given these qualities, it was not readily obvious why IL-2 ultimately failed to meet high expectations therapeutically, although a deeper appreciation for the pleotropic nature of this cytokine has provided some insight. IL-2 binds with intermediate affinity to the heterodimeric IL-2 receptor composed of the IL-2Rβ chain (CD122) and the common gamma chain (γc). In contrast, IL-2 has a much higher affinity for the trimeric receptor that includes IL-2Rβ, γc and IL-2Rα (CD25) [13], which is induced on antigen-activated T- cells and constitutively on FoxP3+ Tregs [14]. Consequently, there is potential for IL-2 to boost effector T-cell responses and even circumvent established anergy, but also to mediate tolerance through the expansion of peripheral Tregs [15]. Therefore, the greatest success for IL-2 immunotherapy has come when IL-2 is used to rescue and expand previously unresponsive tumor-reactive CD8+ T cells in vitro [16,17], and then as a companion therapy to support adoptive transfer of these T cells into lymphodepleted patients where endogenous Tregs numbers are minimized [18]. Such adoptive T-cell immunotherapy is discussed in more detail later in this review.

The use of high-dose IL-2 monotherapy remains mainstream in the treatment of melanoma and renal carcinoma, with more than 100 current clinical trials aimed at defining optimal dosing regimens, and evaluating IL-2 in combination with other immuno-/chemo-therapeutic approaches. Parallel to these clinical studies, efforts are now being made to improve the efficacy of IL-2 treatment by increasing in vivo half-life and enhancing the ability of administered IL-2 to selectively influence desired immune components. For example, association of recombinant IL-2 cytokine with particular anti-IL-2 monoclonal antibodies (mAbs) was shown to form IL-2/mAb complexes that markedly enhance the activity of IL-2 in vivo. Depending on the anti-IL-2 mAbs used, these complexes favor binding to either CD8+ effector T-cells or Tregs [19]. Variants of IL-2 known as ‘superkines’ are also being designed with the ability to preferentially target tumor microenvironment (ALT-801, GA504) and/or activate effectors over regulatory T cells [20]. These IL-2 variants are often fused to antibodies or soluble T-cell receptor (TCR) domains specific for tumor proteins/antigens and can be engineered to have enhanced binding to IL-2Rβ in the absence of CD25 [21]. It will be interesting to observe if these antibody-cytokine complexes or supercytokines translate into better clinical outcomes with higher safety profiles in human patients. Representative clinical trials for IL-2 based therapies are provided in Table 1.

Table 1.

Cytokines.

Cytokine Uses Agent Developmental stage and clinical trial
references
IL-2 Monotherapy IL-2, proleukin,
aldesleukin
Approved for IV infusion in advanced
Renal cell cancer and melanoma
Phase II clinical trials (dosing)
1. NCT01702896– melanoma
2. NCT01702909– kidney cancer
Companion
therapy to
Lymphodepletion
(LD)/ACT/DC
Vaccination/lpi
IL-2, proleukin,
aldesleukin
Phase II clinical trials for melanoma
1. NCT00338377 – LD + ACT + DC Vax
2. NCT01995344–TIL + CTX
3. NCT01701674 – LD + ACT + Ipi
4. NCT02027935 – ACT + CTX + Ipi
Engineered IL-2
variants
RO6895882
(GA504/CEA-IL2v)
Phase I clinical trial
1. NCT02004106–solid tumors
ALT-801 (soluble
P53TCR-IL-2 fusion
protein)
Phase I/II clinical trial
1. NCT01670994– myeloma
IL-2 superkine (super-2) Preclinical animal studies
IL-2/mAb immune
complexes
mlL-2/anti-IL-2,
rh-IL-2/IL-2Rα
Preclinical animal studies

IL-15 Monotherapy rhlL-15 Phase 1 clinical trials
1. NCT01572493 –carcinoma, lymphoma
2. NCT01727076 – solid tumors
3. NCT01021059§ – melanoma or RCC
Companion
therapy(with NK
cell or TIL transfer)
rhlL-15 Phase I clinical trials
1. NCT01385423 –AML
2. NCT01875601 –solid tumors
Phase I/II clinical trial
1.NCT01369888 – melanoma
IL-15/mAb immune
complexes
ALT-803
(IL-15N72D + IL-15RαSu/
Fc)
Phase I/II clinical trials
1. NCT01946789– melanoma
2. NCT01885897 – hematologic malignancy

IL-21 Monotherapy rh-IL-21 Phase I clinical trial
1. NCT00095108§ – melanoma or kidney
cancer
Phase II clinical trial
1. NCT00514085§– melanoma
Combination
therapy (with Ip or
nivolumab)
rh-IL-21 Phase I clinical trials
1. NCT01489059–melanoma
2. NCT01629758–solid tumors

Representative trials, not meant to be exhaustive list of all trials.

Clinical trial status obtained March 2014 from the ClinicalTrials.gov website maintained by the NIH.

Recruiting.

Not yet recruiting.

§

Completed.

Active, not recruiting.

ACT: Adoptive cell transfer; AML: Acute myeloid leukemia; CEA: Carcinoembryonic antigen; CTX: Chemotherapy; DC: Dendritic cell; Ipi: Lipilimumab; IV: Intravenous; LD: Lymphodepletion; mAb: Monoclonal antibody; mIL: Mouse interluekin; RCC:Renal cell cancer; TCR: T-cell receptor; TIL: Tumor infiltrating lymphocytes; rbIL: Recombinant human interluekin; Vax: DC vaccination.

IL-15 & IL-21

Advances in our understanding of the cellular and molecular biology of IL-2 and its receptor complex have provided rationale to better exploit IL-2 signaling pathways to expand and activate T cells in patients with cancer. One such approach is the use of other common-gamma chain cytokines, such as IL-15 and IL-21, which share signaling pathways and immune-modulating activities with IL-2 [22], but may be superior to IL-2 in overcoming mechanisms of tolerance to generate antitumor CD8+ T-cell responses [23,24].

The IL-15 receptor shares the IL-2Rβ and γc chain signaling complex with IL-2, but contains a unique IL-15Rα chain that confers selectivity for IL-15. Differences in the distribution and regulation of the unique α chains of the IL-2R and IL-15R, as well as distinct modes of activity largely account for the proposed advantages of using IL-15 over IL-2 in immunotherapy [25]. IL-15 is currently being tested in 14 clinical trials listed on ClinicalTrials.gov as a recombinant cytokine or DNA vaccine to treat a wide range of advanced solid tumors. Its also being examined as a combination therapy to support adoptively transferred T-cells and NK cells in cancer patients. In similar strategies described above for IL-2, boosting the in vivo efficacy of IL-15 has been explored by creating a complex consisting of the IL-15 cytokine and a soluble form of the IL-15Rα chain [26]. More recently these efforts have evolved toward the creation of a dimeric IL-15 receptor fusion protein (αSu/Fc) complexed with a super agonistic IL-15 mutant cytokine, collectively known as ALT-803 [27]. This IL-15 complex induced robust T-cell and NK cell responses in vivo, as well as antitumor activity in a mouse model of multiple myeloma [28]. ALT-803 is now in Phase I/II clinical trials for treatment of metastatic melanoma (NCT01946789) and relapsed hematologic malignancy (NCT01885897).

Another member of the γc cytokine family, IL-21 is produced by CD4+ and NK T cells, and binds to a heterodimeric receptor composed of the γc chain and a distinct IL-21Rα chain expressed on CD4+ and CD8+ T-cells, as well as other immune cell populations [29]. IL-21 has the potential to impede T-cell tolerance by synergizing with IL-15 to promote activation and proliferation of both memory and naive CD8+ T-cells [30], and by shielding CD8+ T cells from Tregs suppression [31]. Further, IL-21R signaling inhibits Foxp3 expression and attenuates Tregs activity [32,33], representing an important therapeutic advantage over both IL-15 and IL-2. Indeed, IL-21 is being evaluated in clinical trials as a monotherapy for a wide range of cancers, and in combination modalities with more conventional chemotherapies and other immune modulators like anti-PD-1 (NCT01629758), anti-CTLA-4 (NCT01489059). Table 1 provides an overview of clinical trials with γc cytokines.

Costimulation & vaccination

The induction of potent and long-lasting CD8+ T-cell responses is a key goal of therapeutic tumor vaccination. Vaccines are designed to overcome mechanisms of tolerance to tumors by providing antigen, costimulation, or both to awaken tumor-specific T cells. Multiple approaches have been developed to augment the immune response to cancer, ranging from general immune stimulants, such as live attenuated vaccines, to molecularly targeted therapeutics designed to trigger specific activating receptors on immune cell subsets. Here we discuss these approaches and the trials that are underway to evaluate their potential in human patients.

Costimulatory agonists

T-cell-mediated rejection of tumors requires signals from the TCR and costimulatory molecules to activate tumor-reactive T cells, promote differentiation, and induce effector functions. Targeting costimulatory molecules for immunotherapy is based on the premise that tumor-specific T cells exist within a cancer-bearing host, but either fail to recognize tumor antigens in an activating context or have been rendered functionally tolerant/anergic by antigen encounter in the absence of appropriate costimulation. By providing these signals therapeutically, it is expected that T-cell numbers and function will be enhanced, leading to better antitumor immunity. Table 2 provides an overview of these clinical efforts.

Table 2.

Costimulation.

Target Uses Agent Developmental stage and clinical trial references
CD28 Agonistic antibody TGN1412 (CD28-
SuperMAB)
Discontinued due to toxicity
Signaling domain of CAR T-cells CD19.CAR-CD28Z
T-Cells
Phase I clinical trials for CD19+
malignancies
1. NCT02050347
2. NCT02051257
3. NCT01815749

OX40 Agonistic antibody (combination
therapies)
Anti-OX40 mAb Phase I clinical trial
1. NCT01644968 – cancers
Phase I/II clinical trials
1. NCT01303705– prostate cancer
2. NCT01862900– breast cancer
Signaling domain of CAR-T cells iC9-GD2-CD28-
OX40(iC9-GD2)
T cells
Phase I clinical trials
1.NCT01822652–neuroblastoma
2. NCT01953900– sarcoma

4-1BB Agonistic antibody Urelumab
(BMS-663513)
Phase I clinical trial
1. NCT01471210 – solid tumors/NHL
Phase II clinical trial
1. NCT00612664§ – melanoma
Agonistic antibody (combination) PF-05082566 Phase I clinical trial
1. NCT01307267– NHL
Signaling domain of CAR-T cells CD19 scFv
TCR:41BB
Phase II clinical trials
1. NCT02030847 – B-cell ALL
2. NCT01029366§ – B-cell leukemia/lymphoma

CD40 Agonistic antibody(combination) Lucatumumab
(HCD122)
Phase lb clinical trial
1. NCT01275209§ –CD40+follicular lymphoma
Agonistic antibody Dacetuzumab
(SGN-40)
Phase I clinical trial
1. NCT00079716§ – myeloma
Phase I/II clinical trial
1. NCT00283101§ –CLL
Agonistic antibody (combination) CP870-893 Phase I clinical trials
1. NCT0113635 –melanoma
2. NCT00607048§ – solid tumors
Chimeric Agonistic Ab Chi-Lob 7/4 Phase I clinical trial
1. NCT01561911 – neoplasms, lymphoma

Representative trials, not meant to be exhaustive list of all trials.

Clinical trial status obtained March 2014 from the ClinicalTrials.gov website maintained by the NIH.

Recruiting.

Not yet recruiting.

§

Completed.

Active, not recruiting.

ALL: Acute lymphoblastic leukemia; CAR: Chimeric antigen receptor: CLL: Chronic lymphocytic leukemia; mAb: Monoclonal antibody NHL: Non-Hodgkin’s lymphoma.

CD28

The first costimulatory molecule to be discovered and engaged with a mAb to boost T-cell responses against tumor cells was CD28 [34]. Enthusiasm for targeting CD28 therapeutically grew when a CD28 super agonistic antibody (TGN1412) was found to possess strong immunomodulatory properties in pre-clinical models, from the promotion of regulatory T-cell activation to the stimulation of cytotoxic T-cell responses against B-cell leukemia without evoking worrisome proinflammatory mediators. Unfortunately, in a Phase I evaluation in 2006, TGN1412 induced cytokine storm and subsequent multiorgan dysfunction in all healthy volunteers that had received the antibody [35]. Thus, anti-CD28 is not currently being investigated as a therapeutic agent to stimulate T-cell tumor immunity in patients. Indirectly though, treatment with the CTLA-4 antibody, ipilimumab, achieves this objective by blocking ligation of the CD28 homolog and co-inhibitory molecule CTLA-4, which normally outcompetes CD28 for B7 ligand binding, effectively making B7 more available for CD28 costimulation [36]. Additionally, agonistic clinical grade CD28 antibody is used for ex vivo expansion of tumor-reactive CD8+ T-cells and TIL for adoptive cellular therapy [37], and the signaling domain of CD28 has been engineered into newer CAR-based cancer therapies [38], all of which are described later in this review.

OX40

OX40 (CD134) represents a transient activation marker on CD4+ and CD8+ T-cells that is upregulated by TCR engagement and provides co-stimulatory signals to the cells upon engagement of its ligand, OX40L [39,40]. OX40L is predominantly found on activated antigen presenting cells (B cells, dendritic cells, macrophages), but is also expressed on smooth muscle, endothelium, and activated T-cells [41]. Ligation of OX40 promotes T-cell proliferation, survival and effector function [42,43]. Importantly, OX40 signaling has also been reported to overcome CD8+ T-cell tolerance in animal models of cancer [44]. This is due not only to T-cell costimulation, but also the ability to impair Tregs suppressor function [45]. The presence of OX40+ T cells in human malignancies prompted evaluation of agonistic OX40 antibodies clinically [46]. In a Phase I human trial, agonistic antibody was well-tolerated, and enhanced T-cell activation and proliferation while leading to regression of at least one metastasis in 40% of patients receiving a single course of the therapeutic [47]. Five additional clinical studies have been initiated to evaluate the efficacy of OX40 agonistic antibodies either alone or in conjunction with radiation and chemotherapy for prostate cancer (NCT01303705) and breast cancer (NCT01862900),or with checkpoint blockade (ipilimumab: anti-CTLA-4) for metastatic melanoma (NCT01689870). The signaling domain of OX40 is also being incorporated into CAR-expressing T cells for adoptive cell therapy trials to treat neuroblastoma (NCT01822652) and advanced sarcomas (NCT01953900).

4–1BB

4–1BB (CD137) is widely expressed on activated T-cells, NK cells, and other hematopoietic cells, as well as some tumor endothelia [41]. Engagement of 4–1BB with its ligand, 4–1BBL, on activated APC, increases the proliferation of T-cells and their expression of the anti-apoptotic proteins, Bcl-2 and Bcl-xL, promoting their survival [41,48]. In preclinical models, administration of a 4–1BB agonistic antibody reverses CD8+ T-cell tolerance and can promote tumor regression, primarily via its actions on CD8+ T-cells [4951]. For clinical purposes, the broad distribution of 4–1BB creates concerns regarding toxicity. A Phase I clinical trial using the agonistic 4–1BB antibody BMS-663513 for treatment of metastatic melanoma showed signs of immune stimulation and appeared to be well-tolerated, stabilizing disease in 17% of patients for up to 6 months (NCT00309023) [52]. However, a Phase II trial evaluating the same antibody reported severe hepatitis at the highest doses (NCT00612664). A newer 4–1BB antibody, PF05082566, is now in Phase I trials as a monotherapy or in combination with rituximab for non-Hodgkin’s lymphoma (NCT01307267). Like CD28 and OX40, the signaling domain of 4–1BB is being genetically engineered into CAR-expressing T cells for adoptive immunotherapy in multiple cancers, and is discussed later in this review.

CD40

CD40 is broadly expressed, including on professional APC and tumor cells. CD40 ligation stimulates APC maturation and subsequent priming of CD40L-expressing CD4+ T-cells, which then aid in the orchestration of a cytotoxic T-lymphocyte response [53,54]. Thus, targeting CD40 is predominantly an indirect mechanism for promoting immunostimulation of tumor-specific CD8+ T-cells. Agonistic CD40 antibodies have been shown to rescue tolerized cytotoxic lymphocyte responses to poorly immunogenic tumors and have demonstrated potent preclinical antitumor efficacy [5456]. Several different agonistic antibodies have been studied to treat human hematologic malignancies, including lucatumumab (HCD122) and dacetuzumab (SGN-40), but responses were minimal with some toxicity [5760]. For advanced solid tumors, Phase I safety and dose escalation trials have been completed using anti-CD40 (CP870–893). In the first trial, CP870–893 was found to be well-tolerated, with an objective response rate of 14%, and 24% of patients achieving stable disease [61]. Two additional trials were recently completed evaluating CP870–893 in combination with checkpoint blockade (tremelimumab: anti-CTLA-4) (NCT01103635) for late-stage melanoma and in a dose-escalation Phase I trial with the chemotherapeutics, paclitaxel and carboplatin, for solid malignancies (NCT00607048). There is also an active study with the chimeric (mouse and human derived) monoclonal CD40 agonistic antibody Chi Lob 7/4 in Europe (NCT 01561911).

DC vaccines

Dendritic cell (DC) vaccination strategies against cancer exploit the ability of DC to capture, process and present antigens to T-cells, and induce tumor-specific CD8+ T cells to reject tumor and provide long-lived memory [62]. The maturation status of the DC dictates whether tolerance or immunity is induced in engaged T cells [63]. Mature, antigen-loaded DC produce greater amounts of immune-modulating cytokines like IL-12 and IL-15 [25,64], and upregulate expression of co-stimulatory molecules required to enhance effector differentiation of CD8+ T cells and overcome tumor-associated tolerance [50,56,65]. To generate mature DC for vaccination, patient-derived DC are cultured with adjuvant, or alternatively, monocytes are differentiated with cytokines (e.g. GM-CSF and IL-4) [66]. Clinical studies have demonstrated that ex vivo generated DC vaccines are safe and capable of inducing the expansion of circulating tumor-specific T-cells [67]. Recently, Sipuleucel-T (Provenge) became the first commercial DC vaccine approved by the US FDA for treatment of prostate cancer (Table 3). This personalized vaccine is created through enrichment and activation of patient-derived DC by culturing with GM-CSF and the prostate cancer antigen, prostatic acid phosphatase (PAP). Sipuleucel-T treatment induces T-cell proliferation and PAP-specific immune responses, and provides a 4-month survival benefit to vaccinated patients [68]. Additional DC vaccines (DCVax-L) are now being commercially developed and tested in clinical trials against glioblastoma (NCT00045968), renal carcinoma (NCT01582672), ovarian cancer (NCT00683241), and other solid tumors (NCT01882946).

Table 3.

Vaccination.

Target Strategy Agent Developmental stage and clinical trial
references
Dendritic cells Ex vivo generation Sipuleuel T
(ACP8015/
Provenge)
US FDA approved therapeutic cancer vaccine
DC Vax-L Phase I, I/II clinical trials
1. NCT00683241– ovarian cancer
2. NCT01882946– solid tumors
Phase III clinical trials
1. NCT00045968– glioma
2. NCT01582672 – RCC
In situ vaccination CDX-1401 (DEC-
205/NYESO-1)
Phase 1 clinical trials
1. NCT01522820 – solid tumors
2. NCT01834248 – AML/MDS
DC tumor cell fusion
vaccines
Phase I clinical trial
1. NCT00459069 – myeloma
Phase I/II, II clinical trials
1. NCT00799110§ – ovarian cancer
2. NCT00622401§ – breast cancer
Combination
therapy
Sipuleucel-T
+ ipilimumab
(CTLA-4
blockade)
Phase I clinical trial
1. NCT01832870§ – prostate cancer
Phase II clinical trial
1. NCT01804465§ – prostate cancer
DC tumor
fusion vaccine +
pidilizumab (PD1
blockade)
Phase II clinical trials
1. NCT01441765–RCC
2. NCT01096602–AML
3. NCT01067287– myeloma

Listeria
monocytogenes
Live-attenuated
Listeria encoding
LLO-E7 fusion
protein
Lovaxin-C
(ADXS-11-001,
Lm-LLO-E7)
Phase I clinical trial
1. NCT01598792 – oropharyngeal cancer
Phase II clinical trials
1. NCT01116245–CIN
2. NCT01266460– cervical cancer
Live-attenuated
Listeria expressing
human mesothelin
CRS-207 Phase I clinical trial
1. NCT01675765–pleural mesothelioma
Phase II clinical trial
1. NCT02004262 – pancreatic cancer
Live-attenuated
Listeria expressing
EGFRvlll and
NY-ESO-1
ADU-623 Phase I clinical trial
1. NCT01967758 – astrocytoma, glioblastoma

Representative trials, not meant to be exhaustive list of all trials.

Clinical trial status obtained March 2014 from the ClinicalTrials.gov website maintained by the US NIH.

Recruiting.

Completed.

§

Active, not recruiting.

AML: Acute myeloid leukemia; CIN: Cervical intraepithelial neoplasia; DC vax: Dendritic cell vaccination; LLO: Listeriolysin; MDS: Myelodysplastic syndrome; RCC: Renal cell carcinoma.

A related therapeutic approach is in situ DC vaccination. Here, tumor antigen is delivered to endogenous DC by coupling antigens to antibodies specific for DC surface receptors such as DEC205 [69]. Receptor binding leads to antigen internalization, processing, and presentation by endogenous DC matured via a co-administered adjuvant. Two Phase I trials have been initiated to investigate vaccination with a DEC-205/NY-ESO-1 fusion protein for vaccination of patients with a broad range of cancers including acute myeloid leukemia or myelodysplastic syndrome [NCT01834248], or with sirolimus in a variety of solid tumors [NCT01522820]. DC vaccines are also being explored in combination with other immune modulating treatments like checkpoint blockade (Table 3). The combination of MART-1 peptide-pulsed DC and the CTLA-4 blocking antibody Tremelimumab increased MART-1 specific CD8+ T cells and achieved objective response in 25% of melanoma patients [70]. A Phase II study using PD-1 blockade in conjunction with the DC/Renal Cell Carcinoma Fusion Cell Vaccine is now recruiting patients (NCT01441765).

Listeria monocytogenes

A variety of live attenuated bacterial and viral vectors are being explored as anticancer agents, with Listeria monocytogenes ranking among the most well-characterized for therapy. Its intracellular lifestyle and natural targeting of DC and other professional APC makes Listeria particularly adept at eliciting the CD8+ T-cell-mediated immunity required to effectively eliminate tumor [71]. The ability to manipulate the Listeria genome to encode desired antigens or adjuvant proteins that are secreted in situ in infected cells helps drive tumor-specific responses, including reinvigoration of tolerant self-tumor-reactive CD8+ T-cells [72,73]. Furthermore, the inflammatory responses driven by Listeria vaccines efficiently promote tumor infiltration by activated CD8+ T-cells and reduce suppressive cell populations [74].

Attenuated strains of L. monocytogenes being assessed for the clinical treatment of cancer have demonstrated an acceptable safety profile and provided early indications of activity. Lovaxin C (ADXS-11–001, LM-LLO-E7) encodes the Human Papilloma Virus (HPV) E7 antigen fused to Listeriolysin (LLO), an antigenic virulence factor that enhances the immunogenicity of the E7 protein [75]. In a Phase I/II study for advanced cervical cancer, 53% of patients had stable disease and 31% of patients had a reduction in tumor size. [76]. These promising results have lead to Phase II trials for cervical intraepithelial neoplasia (NCT01116245), cervical carcinoma (NCT01266460), and Phase I trials for HPV16+ oropharyngeal cancer (NCT01598792). Similarly, when the mesothelin antigen encoding CRS-207 (Listeria ΔactA/ΔinlB-mesothelin) was evaluated in patients with mesothelin-positive malignancies, 35% of patients survived greater than 15 months and 83% of these individuals mounted an LLO-specific T-cell response [77]. Subsequent clinical trials with CRS-207 have been initiated in conjunction with GVAX for metastatic pancreatic cancer (NCT01417000, NCT02004262), or with pemetrexed and cisplatin for pleural mesothelioma (NCT016757650). A Phase I trial examining a newer agent, ADU-623, for glioblastoma multiform (NCT01967758) was recently initiated (Table 3).

Adoptive cell transfer

Adoptive cell transfer (ACT) strives to overcome tolerance by generating large numbers of appropriately activated T cells outside of the immunosuppressive host environment and re-infusing them back into patients where they can mediate tumor regression [37]. Early trials of this approach were limited by their inability to achieve engraftment of large numbers of high affinity tumor-specific T cells, but strategies such as lymphodepletion, elimination of specific suppressor cell subsets, and genetic engineering have enabled adoptive cell-based therapies to achieve dramatic tumor regression in human patients [78].

Genetically engineered T-cell receptors

T-cell receptor (TCR) engineering is a strategy to overcome the obstacles of limited numbers of peripheral tumor-antigen reactive T-cells and the restriction of lower affinity TCR expression. This is accomplished by introducing genes encoding a high affinity TCR to redirect the specificity of patient-derived peripheral T-cells [79]. Such redirected T-cells have specificity for tumor-associated antigens (TAA), and can be re-infused into patients to provide tumor immunity. The first clinical application of genetically engineered TCR for adoptive immunotherapy utilized a MART-1-specific TCR and reported melanoma tumor regression and durable objective responses in two out of 15 patients with rapidly progressive and therapy-resistant metastatic melanoma [80]. These proof of concept studies have led to several subsequent trials, including ongoing Phase II trials for MART-I (NCT00910650) and NY-ESO-1 (NCT01697527, NCT00670748) specific TCR in patients with melanoma or other advanced solid malignancies (Table 4). One limitation of this approach has been the reliance on TCR derived from endogenous TAA-reactive T cells, which are not always high affinity. This is being countered by introducing mutations in the TCR complementarity determining regions, enhancing TCR affinity several orders of magnitude [81]. Affinity-enhanced TCR directed toward the cancer testis antigen, NY ESO-1, were found to be both safe and effective [82], and are the focus of two current clinical trials (NCT01892293 and NCT01967823). However, there is accumulating evidence of severe off-target toxicity with affinity-enhanced TCR specific for other targets, such as MAGE-A3 [83,84], suggesting the future of this strategy will rely on better toxicity screening. Extensive reviews covering the challenges and advances in TCR gene therapy have been provided elsewhere [79,85].

Table 4.

Engineered T-cell receptors/chimeric antigen receptors.

Target antigen Structure/endodomain Agent Developmental stage and clinical trial references
TCR

MART-1 T-cell receptor MART-1-specific TCR Phase II clinical trials in melanoma
1. NCT00706992#
2. NCT00509288§
3. NCT00910650

NYESO-1 NY ESO-1-specific TCR Phase I/II clinical trials in myeloma
1. NCT01352286
2. NCT01892293
Phase II clinical trials in NY-ESO-1 + cancers
1. NCT00670748
2. NCT01697527
3. NCT01967823

MAGEA3 PG13-MAGE-A3 TCR9W11
(MAGE-A3/12TCR)
Phase I/II clinical trial
1. NCT01273181# – MAGEA3/12+ cancer

GP100 gp100-specific TCR Phase II clinical trial
1. NCT00509496# – melanoma

CAR

CD19 CD28-CD3ζ CD19CAR-28-zetaT cells Phase I, I/II clinical trials
1. NCT00466531–CD19+CLL/BCL
2. NCT00586391 – B-NHL, ALL & CLL
41BB-CD3ζ CART-19 TCR zeta:4-1BB
(CTL019)
Phase II clinical trials
1. NCT02030834 – CD19+lymphomas
2. NCT02030847 – B cell ALL
3. NCT01029366§ – B cell leukemia/lymphoma
4. NCT01747486 – CD19+CLL or SLL

CD20 CD28-41BB-CD3ζ CART-20 TCR zeta:4-1BB Phase I clinical trials in B- cell leukemia/lymphoma
1. NCT00621452
2. NCT01735604

CEA CD28-CD3ζ Anti-CEA Designer T-cells Phase II clinical trials
1. NCT01723306 – adenocarcinomas

Her2 CD28-CD3ζ HER.CAR CMV-specific CTLs Phase I clinical trials
1. NCT01109095 – GBM
2. NCT00889954 – HER2+ cancer

GD2 CD28-OX40-CD3ζ iC9-GD2-CD28-OX40
(iC9-GD2) T-cells
Phase 1 clinical trials
1. NCT01822652 – neuroblastoma
2. NCT01953900 – sarcoma

Representative trials, not meant to be exhaustive list of all trials.

Clinical trial status obtained March 2014 from the ClinicalTrials.gov website maintained by the US NIH.

Recruiting.

Not yet recruiting.

§

Completed.

Active, not recruiting.

#

Terminated.

ALL: Acute lymphocytic leukemia; CAR: Chimeric antigen receptor: CLL: Chronic lymphocytic leukemia; GBM: Glioblastoma multiforme; NHL: Non-Hodgkin’s Lymphoma; SLL: Small Lymphocytic Lymphoma; TCR: T-cell receptor.

Chimeric antigen receptors

A promising new approach to prevent tolerance in autologous adoptively transferred tumor-reactive T-cells is expression of genetically engineered chimeric antigen receptors (CAR). Unlike engineered TCR, CAR molecules are completely artificial receptors composed of an antibody subunit capable of binding specific tumor antigens independent of MHC, a trans-membrane domain, and cytoplasmic domains that transduce activation signals to the T-cell upon antigen encounter [38]. Utterly simplistic in concept, the cytoplasmic tails can be tailored to contain signaling domains from costimulatory molecules (e.g., CD28 and 4–1BB) such that antigen recognition is always associated with powerful costimulation [86]. T-cells that express tumor antigen-specific CAR are considerably less vulnerable to mechanisms of tumor evasion and immune suppression (i.e., tolerance) due to this high level of consistent activation. In addition, tumor recognition by CAR is not compromised by alterations in tumor MHC expression or epitope processing, or by TCR complex dissociation on responding T-cells. Furthermore, the antibody-antigen interaction is often of much higher affinity compared with natural TCR binding to MHC/peptide. The fact that CAR do not rely on MHC/peptide-binding carries the added advantage that they are not restricted by a patients’ HLA type.

The primary limitation of CAR-mediated immunotherapy is that antigens can only be recognized on the surface of tumors, leaving intracellular tumor-antigens essentially undetectable. However, this is not a serious drawback for those cancers that express well-known and targetable surface antigens, like CD19 on B cell-derived leukemia and lymphoma. T-cells expressing CD19-specific CAR have shown remarkable efficacy at clearing B-cell tumors in animal models [86], and proven clinically promising in human patients [87]. That being said, identification of novel targetable antigens will be key for CAR-based immunotherapy moving forward. This is aided by the antibody–antigen recognition by CAR molecules, which open up a wider range of targets that are not typically involved in TCR-mediated tumor cell recognition, such as carbohydrates and glycolipid tumor antigens [88]. There are approximately 40 studies listed on [89] designed to explore CAR-expressing T cells for immunotherapy in human cancer patients (Table 4).

The next generation of CAR-expressing T cells is being designed in laboratories and tested in animal models. Such T cells are referred to as ‘TRUCK’s (T-cells redirected for universal cytokine killing), and express CAR along with a separate transgene to allow expression of secreted factors like cytokines [90]. This concept extends the capabilities of CAR-expressing T-cells beyond tumor antigen recognition and direct cytotoxicity, and brings to bear the potential to direct other immune responses within the tumor environment. For example, two reports have demonstrated that IL-12 expression by tumor-specific CAR-T-cells were more effective at clearing detectable tumors than the same CAR-T-cells that lacked IL-12 expression [91,92]. Moreover, Chmielewski et al. showed that IL-12 secretion by these CAR-T-cells even promoted in vivo killing of antigen-negative tumors not recognized by the specific CAR being expressed. This is a particularly important enhancement of the CAR concept, as antigen-loss tumor variants represent a real impediment to CAR-based immunotherapy in patients [87]. This compelling result demonstrates the immunomodulatory power of IL-12, a pleiotropic cytokine with the ability to induce expression of the costimulatory ligands CD80 and CD86 on surrounding cells, promote localized Th1 responses (including IFN-γ secretion), recruit natural killer cells and macrophages, and provide resistance to the suppressive effects of Tregs and myeloid derived suppressor cells (MDSCs) [93].

It is tempting to speculate that the bright future of CAR and TRUCK-based immunotherapies for cancer will be limited only by the number of antigens that can successfully be targeted. However managing the risks associated with unleashing such a robust immune response in patients is also key to the future success of these treatment strategies. Engineered T cells can cause autoimmunity, induce cytokine storms, and any genetic modifications can lead to insertional mutagenesis, causing severe toxicity and even death in patients [87,9495]. Some of these events could potentially be reversed by selective deletion of the engineered T cells themselves, and this is being explored by introducing a ‘suicide gene’ along with other CAR to provide such an option for patients [96].

Lymphodepletion

Host conditioning or ‘lymphodepletion’ prior to adoptive cell transfer provided one of the earliest leaps forward for ACT immunotherapy. A lymphopenic environment promotes T-cell homeostatic proliferation, effector function, memory-like differentiation, and even transient rescue of CD8+ T-cell tolerance [9799]. In preclinical models, lymphodepletion via total body irradiation (TBI) and/or cytotoxic drugs prior to ACT were found to enhance immune responses to cancer [100,101]. Similarly immunodepleting chemotherapy in human cancer patients prior to T-cell infusion led to the first reported tumor regressions by ACT, with 47–51% objective responses achieved in two separate melanoma trials [18,102]. Several mechanisms by which this disruption of immune homeostasis might overcome tolerance to provide better anticancer immunity have been described, including the creation of space in the lymphoid compartment for low-avidity tumor-reactive clones to expand or for adoptively transferred cells to engraft, the depletion of suppressor cell populations, and the elimination of competition for homeostatic cytokines [103].

The success of patient preconditioning has prompted recent efforts probing the extent of lymphodepletion necessary to improve patient outcomes. Lymphodepletion by chemotherapy alone or in combination with TBI were compared in sequentially treated cohorts of melanoma patients [104]. Here, 40% complete responses and 72% objective responses were observed for highest dose TBI (12 Gy), highlighting the critical role for host manipulation in optimizing ACT. These results are not achieved without consequence, as some autoimmunity toward healthy melanocytes and other toxicities do occur, but are generally well tolerated. Currently, there are 25 clinical trials employing lymphodepletion in combination with other approaches such as ACT, DC vaccines, systemic IL-2, or molecularly targeted therapeutics. Fifteen of these trials are for metastatic melanoma, while others are aimed at ovarian (NCT01312376), kidney (NCT00091611), and nasopharyngeal (NCT00078546) neoplasms.

Inhibitory cell depletion

One of the presumed reasons for success of lymphodepletion strategies is the reduction of host suppressor cells. Suppressive cell types can dominate the tumor microenvironment, and the most well-defined of these are the Tregs [105]. Tregs are a population of CD4+ CD25hi Foxp3+ T cells whose role in the maintenance of self-tolerance and immune homeostasis is best demonstrated by the lymphoproliferation and inflammatory disease observed in mice and humans unable to generate these cells [14,106]. The accumulation of Tregs in malignancy is generally associated with a poor prognosis due to local immune suppression. But this is somewhat controversial and the current consensus is that the prognostic value of a high Tregs to effector T-cell ratio is likely dependent on the type of cancer [107]. Thus, in select tumors, depletion of Tregs may provide therapeutic benefit. The first strategies aimed at depleting Tregs targeted their high CD25 expression with monoclonal antibodies (daclizumab) or antibody-conjugated toxins (denileukin diftitox), which reduced Tregs numbers in patients and correlated with enhanced T-cell responses post-vaccination [108,109]. However, subsequent studies have failed to consistently observe these promising results, likely attributable to simultaneous depletion of effector T cells that also express CD25 [110]. To further define their clinical activity, both drugs remain under investigation in numerous studies for treatment of diverse human cancers. Additional methods to preferentially eliminate Tregs for cancer therapy have entered clinical trials, including low metronomic doses of cytotoxic drugs such as cyclophosphamide (NCT01581970, NCT01462214), and anti-CCR4 mAb (KW-0761, mogamulizumab) (NCT01929486). Finally, recent studies suggest that the antitumor activity of CTLA-4 targeted antibodies is achieved in part by depletion of Foxp3+ CD4+ Tregs, contributing to the activation of tumor-resident effector T-cells [111,112]. Of note, current therapies targeting Tregs nonspecifically deplete all Tregs, which creates a general risk of autoimmunity in patients. A key advancement would be the ability to specifically target the Treg-cell subpopulations contributing to tumor progression.

Myeloid-derived suppressor cells (MDSC) are a population of myeloid progenitor cells that fail to fully differentiate into DC, macrophages, or granulocytes. They are collectively identified by their suppressive phenotype and expression of the myeloid markers, Gr1 or CD11b in mice, or CD33+CD11b+HLA-DRlow/neg in humans [113]. Suppression of CD8+ T cells by MDSC can be achieved by many different mechanisms, including downregulation of the TCRζ, proliferative arrest induced by depletion of amino acids, and altered migration via cleavage of CD62L by the ADAM17 ‘sheddase,’ which have been described in more detail elsewhere [114,115]. Tumor-derived factors promote the accumulation of MDSC in both blood and tumor of cancer-bearing hosts, and frequency inversely correlates with prognosis [116]. A wide range of therapeutic targets have been identified for eliminating MDSC, from preventing their formation and function, to depleting them or inducing their differentiation, which have been reviewed by others [117]. Some of these approaches are in clinical trials, employing therapeutics already FDA-approved for other indications, such as Tadalafil (a PDE5 inhibitor), which is being evaluated in head and neck cancer (NCT01697800). For depletion of MDSC, cytotoxic therapy with gemcidibine is being evaluated in a Phase I trial for sarcoma (NCT01803152).

Checkpoint blockade

The fate of any responding T-cell is largely determined by a balance between positive (co-stimulatory) and negative (co-inhibitory) signals. Several receptor-based pathways have been identified as negative regulators or ‘checkpoints’ of T-cell activation, which can induce tolerance or dysfunction in tumor-reactive T-cells. Overcoming tolerance induced by these receptors has recently been transformed from proof-of-principle experiments in laboratories to clinically effective treatments for patients with cancer. The concept of blocking negative regulatory receptors using specific monoclonal antibodies is referred to collectively as checkpoint blockade immunotherapy. Here, we discuss the molecular mechanisms that underlie this potent new therapeutic strategy, and review results from recent clinical trials.

CTLA-4

(CTLA-4; CD152) is expressed on activated CD4+ and CD8+ T cells, and on Tregs. Its functions in T-cell biology, during immune responses to infection, and as a target for cancer immunotherapy have been well described and reviewed elsewhere [118]. CTLA-4 is a homologous counterpart to the co-stimulatory receptor CD28, both of which bind to CD80 and CD86 on APCs. The importance of CTLA-4 for immune tolerance is clear [119], but its role is complex, involving several overlapping mechanisms that serve to blunt T-cell responses in a range of settings. These include out-competing lower affinity CD28 molecules for ligand binding to minimize T-cell co-stimulation, recruitment of inhibitory phosphatases to the TCR complex to disrupt positive signaling cascades, and removing CD80 and CD86 from the surface of APC by trans-endocytosis, thereby diminishing the ability of APC to properly activate otherwise responsive T-cells [120122].

Based on the criteria above, it is easy to understand why exploitation of the CTLA-4 receptor/pathway is an attractive strategy to modulate T-cell immunity. Indeed, anti-CTLA-4 was the first monoclonal antibody (ipilimumab) to be FDA-approved for checkpoint blockade treatment in cancer patients. There are currently more than 150 ongoing clinical trials investigating the safety and efficacy of CTLA-4 antibodies (ipilimumab and tremelimumab; Table 5, with most focused on boosting immune responses to cancer, predicated on the promising results in melanoma patients [123]. These monoclonal antibodies are thought to work by binding to CTLA-4 on the surface of effector T-cells and blocking natural ligation with B7 molecules. From the perspective of tumor-reactive effector CD8+ T-cells, this prevents negative intracellular signal transduction downstream of CTLA-4, and also results in more unbound B7 available for ligation to CD28 for co-stimulation. On Tregs, there is evidence that CTLA-4 has a direct role in the suppressive phenotype, which may be inhibited with blockade antibody [124,125]. It has also been demonstrated in mice that anti-CTLA-4 binds to CTLA-4high Tregs cells within tumors and promotes antibody-mediated depletion of these Tregs populations while sparing effector T-cells [112]. However, it is important to acknowledge the inevitable flipside of overcoming these tolerizing mechanisms and reinvigorating a powerful immune response, as blockade of CTLA-4 in patients comes at the cost of potentially severe autoimmunity [123]. Predicting and managing these immune related toxicities is vital for the future success of checkpoint blockade immunotherapy.

Table 5.

Individual checkpoint blockade.

Target Agent Antibody type Developmental stage and clinical trial
references
CTLA4 Ipilimumab (MDX-010) Human lgG1 mAb US FDA approved for melanoma
Phase I/II, II clinical trials in melanoma
1. NCT01585194
2. NCT01689974
Phase III clinical trials in melanoma
1. NCT00094653§
2. NCT01515189
Tremelimumab (CP-675, 206) Human lgG2 mAb Phase II clinical trials
1. NCT00471887 – melanoma
2. NCT01649024 – mesothelioma

PD1 Nivolumab
(BMS-936558 or MDX1106)
Human lgG4 mAb Phase 1 clinical trials
1. NCT01176461 – melanoma
Phase I/II clinical trial
1. NCT01928394 – solid tumors
Phase II clinical trial
1. NCT01354431 – renal cell cancer
Phase III clinical trials
1. NCT01844505 – untreated melanoma
Lambrolizumab
(MK-3475)
Human lgG4 mAb Phase I clinical trial
1. NCT01295827 – solid tumors
Phase II clinical trial
1. NCT02085070 – melanoma and NSCLC
Phase III clinical trial
1. NCT01866319 – melanoma
Pidilizumab(CT-011) Human lgG1 mAb Phase I/II clinical trial
1. NCT01952769 – glioblastoma multiforme
Phase II clinical trials
1. NCT01435369§ – melanoma
2. NCT01313416 – pancreatic cancer
AMP-224 PDL2 Ig fusion protein Phase I clinical trial
1. NCT01352884 – cancers

PDL1 MDX-1105
(BMS-936559)
Human lgG4 mAb Phase 1 clinical trial
1. NCT00729664 – solid tumors
MPDL3280A Engineered lgG1
(no ADCC)
Phase 1 clinical trial
1. NCT01375842 – Solid tumors
2. NCT02013219 – NSCLC
Phase II clinical trials for NSCLC
1. NCT02031458
2. NCT01903993

LAG-3 BMS-986016 Human IgG mAb Phase 1 clinical trials
1. NCT0206176 – hematologic neoplasms
IMP321 Soluble LAG-3 Phase I clinical trial
1. NCT00351949§ – renal cell cancer
Phase I/II clinical trial
1. NCT01308294 – melanoma

Representative trials, not meant to be exhaustive list of all trials.

Clinical trial status obtained March 2014 from the ClinicalTrials.gov website maintained by the NIH.

Recruiting.

Not yet recruiting.

§

Completed.

Active, not recruiting.

ADCC: Antibody-dependent cell-mediated cytotoxicity; mAb: Monoclonal antibody; NSCLC: Non-small-cell lung cancer.

PD-1 & PD-L1

Programmed death-1 (PD-1; CD279) is a member of the same family of receptors as CD28 and CTLA-4, and is broadly expressed on lymphoid and myeloid cells [126]. PD-1 binds uniquely to the B7 ligands PD-L1 and PD-L2 on APC and other surrounding tissues, greatly influencing the fate of responding CD8+ T cells in settings of chronic infections and cancer [127129]. On T-cells, PD-1 is expressed after antigen encounter, but acts almost immediately to impede T-cell activation by recruiting the phosphatases SHP-1 and SHP-2 through signaling motifs in the PD-1 cytoplasmic tail, which reduces Akt phosphorylation, and diminishes T-cell metabolism, proliferation and survival [130]. This is particularly relevant to cancer immunology because many different types of tumor cells can express PD-L1 [131].

Efforts to attenuate the PD-1/PD-L1 pathway have followed a similar blueprint as CTLA-4, focusing on antagonistic antibodies that block receptor ligation. Preclinical work in animal models has demonstrated that in vivo administration of such antibodies can restore function in otherwise ineffective T-cells to boost antitumor and antviral responses [127,128]. Three recent clinical trials using different PD-1 or PD-L1 antibodies reported objective responses in 6–38% of patients with different cancer types [132134]. However, treatment related immune toxicities (> grade 3) were reported in 9–14% of patients treated, a similar frequency reported for the anti-CTLA-4 monoclonal antibody, ipilimumab [123]. The autoimmune events associated with blocking PD-1 are similar to those that occur when blocking CTLA-4 and frequently include skin or gastrointestinal manifestations, such as colitis or dermatitis [135]. Fortunately, as more patients are treated with checkpoint blockade immunotherapy, these autoimmune events are becoming easier to predict and manage by inducing immunosuppression, and altering or discontinuing treatment [136]. There are currently more than 50 trials listed on [89] investigating the safety and efficacy of PD-1 pathway blockade for treatment of a wide range of different human cancers (Table 5).

Combination immunotherapy

The exploitation of negative regulatory receptors to overcome or prevent T-cells tolerance has not stopped at CTLA-4 and PD-1. Several others, including LAG-3 and TIM-3, have been shown to influence T-cell dysfunction [137139], and are under investigation as potential checkpoints for cancer immunotherapy intervention, which has been discussed elsewhere in more detail [36,140]. While most checkpoint blockade antibodies have been investigated as monotherapies, there is general consensus that combination strategies are likely to be more effective [36,141]. The logic behind this theory lies in the understanding that each regulatory receptor plays a unique role in T-cell tolerance, and simultaneous blockade of multiple pathways represents a means to restore more immune functions. This notion has been demonstrated in animal models of cancer [128,142144], and recapitulated in melanoma patients treated with CTLA-4 and PD-1 double blockade [145]. Nearly as remarkable as the 53% objective response rate achieved in patients was the realization that adverse immune-related events associated with this combination blockade was similar in nature to individual antibody regimens, and that these were manageable or even reversible in most cases. There are currently at least nine trials listed on ClinicalTrials.gov testing the combination of anti-PD-1 (8 with Nivolumab and 1 with MK-3475) and anti-CTLA-4 (ipilimumab), and one examining anti-LAG-3 (BMS-986016) and nivolumab. Many more are studying checkpoint blockade antibodies in combination with other more conventional chemotherapies (Table 6).

Table 6.

Combination checkpoint blockade.

Target Agent Antibody type Developmental stage and clinical trial
references
CTLA4 + PD1 Ipilimumab
Nivolumab
Phase I clinical trials
1. NCT01024231 – melanoma
Phase I/II clinical trials
1. NCT01928394 – solid tumors
2. NCT02060188 – colon cancer
3. NCT01927419§ – melanoma
Phase III clinical trials
1. NCT01844505 – untreated melanoma

CTLA4 + PD1 Ipilimumab
Lambrolizumab
Phase I/II clinical trials
1. NCT02089685 – melanoma or carcinoma

CTLA4 + PDL1 Tremelimumab+M
EDI4736
Phase I clinical trials
1. NCT01975831 – solid tumors
2. NCT02000947 – NSCLC

PD1 + LAG-3 Nivolumab
BMS-986016
Phase I clinical trial
1. NCT01968109 – solid tumors

Representative trials, not meant to be exhaustive list of all trials.

Clinical trial status obtained March 2014 from the ClinicalTrials.gov website maintained by the NIH.

Recruiting.

Not yet recruiting.

§

Active, not recruiting.

mAb: Monoclonal antibody; NSCLC: Non-small-cell lung cancer.

Conclusion & future perspective

Since the first attempts to harness cellular immunity to eliminate cancer in human patients over 30 years ago, much has been learned about the potential of the immune system to control cancer and how immunotherapy can boost this potential in patients. An appreciation for immune tolerance has aided this process, as we now understand that cancer escapes immune control by failing to elicit sufficient immunity or more actively by inducing peripheral tolerance. Thorough understanding of CD8+ T-cell tolerance has been instrumental to clinical progress, and enabled the design of targeted approaches to achieve antitumor responses by tumor-reactive T cells that otherwise remain ignorant due to low-avidity receptors or anergic by the immuno-suppressive tumor microenvironment. At the same time, advances in biotechnology and molecular techniques have also aided this endeavor, providing a foundation for engineering T cells of desired antigenic specificity. The recent achievements with these approaches for patients with advanced-stage, treatment refractory disease have collectively validated the long-standing idea that immunity is vitally important in the control and elimination of cancer. The paradigm has shifted for treating cancer patients exclusively with tumor-targeted approaches to now include strategies aimed at stimulating immunity. Improving upon these successes will likely require careful incorporation of several treatment modalities in combination approaches, some of which are already being explored in clinical trials. It will also rely on the same ‘bench-to-bedside and back’ translational philosophy that has characterized the development of immunotherapy thus far where observations from animal studies are exploited to provide clinical benefit for human cancer patients, and this success in patients guiding the next steps in preclinical investigation.

Supplementary Material

Supplementrary

Executive summary.

CD8+ T-cell tolerance & cancer immunotherapy

  • CD8+ T cells are the predominant effector in most cancer immunotherapy settings.

  • Tolerance represents a major obstacle to eliciting antitumor CD8+ T-cell responses; thus, immunotherapeutic approaches aim to overcome tolerance.

  • Autoimmunity is a concerning but often manageable risk of cancer immunotherapy.

Cytokines

  • IL-2, IL-15, and IL-21 are capable of overcoming tolerance and promoting CD8+ T-cell immunity, with variants and cytokine-antibody immune complexes representing new tools to potentiate the desirable properties of cytokine therapy.

  • Current clinical trials focus on cytokine dosing regimens and combination approaches with other therapeutics, to define the safety and efficacy of cytokine based therapeutics.

Costimulation & vaccination

  • Costimulatory agonists

    • -

      Ligation of costimulatory receptors (CD28, OX40, 4–1BB, CD40) can prevent the induction of T-cell tolerance and overcome established tolerance.

    • -

      Costimulatory agonistic antibodies are being evaluated as mono/combination therapies for a variety of cancers, and the signaling domains of costimulatory receptors have been incorporated into chimeric antigen receptors (CAR).

  • Dendritic cell (DC) vaccines

    • -

      Mature DC presenting tumor antigens promote T-cell immunity over tolerance.

    • -

      Newer approaches include in situ DC vaccination, DC-tumor fusion vaccines, and combination therapies.

  • Listeria vaccines

    • -

      Listeria vaccines encoding tumor antigens naturally secrete these products into infected dendritic cells that efficiently prime CD4+ and CD8+ T-cell immune responses.

    • -

      Early trials with these vaccines have found them to be safe, immunogenic and capable of reducing tumor burden; additional trials are underway.

Adoptive cell transfer

  • Adoptive cell transfer overcomes tolerance by removing tumor-reactive T cells from the immunosuppressive host, expanding them ex vivo, and reinfusing them back into patients to mediate tumor regression.

  • T-cell specificity can be redirected by transduction of high affinity T-cell receptors (TCR) or CAR to overcome tolerance-imposed limitations on tumor-specific T-cell number, specificity, and avidity.

Checkpoint blockade

  • Inhibitory co-receptors (CTLA-4, PD-1, among others) play a role in the induction and maintenance of T-cell tolerance by transducing negative signals during antigen encounter.

  • Antibody blockade of these receptors, individually and in combination, can restore function in tolerant T-cells and boost antitumor responses.

Conclusion & future perspective

  • A basic understanding of immune tolerance has elucidated key principles guiding the development of cancer immunotherapeutics.

  • Recent success in patients with advanced disease validates the long-standing idea that the immune system can eliminate cancer and is driving new areas of investigation.

Acknowledgments

The authors receive research support from the NIH/National Institute of Allergy and Infectious Disease (R01AI087764) to RM Teague, the Cancer Research Institute (Investigator Award) to RM Teague, and the NIH/National Cancer Institute (F30CA180375) to SR Jackson.

Footnotes

Disclaimer

The content is solely the interpretation of the authors and does not necessarily represent the official views of the NIH.

Financial & competing interests disclosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as:

• of interest; •• of considerable interest

  • 1.Coley WB., II Contribution to the Knowledge of sarcoma. Ann. Surg. 1891;14(3):199–220. doi: 10.1097/00000658-189112000-00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Mitchison NA. Studies on the immunological response to foreign tumor transplants in the mouse. I. The role of lymph node cells in conferring immunity by adoptive transfer. J. Exp. Med. 1955;102(2):157–177. doi: 10.1084/jem.102.2.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hernandez J, Aung S, Redmond WL, Sherman LA. Phenotypic and functional analysis of CD8(+) T cells undergoing peripheral deletion in response to cross-presentation of self-antigen. J. Exp. Med. 2001;194(6):707–717. doi: 10.1084/jem.194.6.707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Redmond WL, Sherman LA. Peripheral tolerance of CD8 T lymphocytes. Immunity. 2005;22(3):275–284. doi: 10.1016/j.immuni.2005.01.010. [DOI] [PubMed] [Google Scholar]
  • 5.Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science. 1976;193(4257):1007–1008. doi: 10.1126/science.181845. [DOI] [PubMed] [Google Scholar]
  • 6.Lotze MT, Robb RJ, Sharrow SO, Frana LW, Rosenberg SA. Systemic administration of interleukin-2 in humans. J. Biol. Response Mod. 1984;3(5):475–482. [PubMed] [Google Scholar]
  • 7.Lotze MT, Matory YL, Ettinghausen SE, et al. In vivo administration of purified human interleukin 2. II. Half life, immunologic effects, and expansion of peripheral lymphoid cells in vivo with recombinant IL 2. J. Immunol. 1985;135(4):2865–2875. [PubMed] [Google Scholar]
  • 8.Cheever MA, Thompson JA, Peace DJ, Greenberg PD. Potential uses of interleukin 2 in cancer therapy. Immunobiology. 1986;172(3–5):365–382. doi: 10.1016/S0171-2985(86)80118-8. [DOI] [PubMed] [Google Scholar]
  • 9.Lotze MT, Chang AE, Seipp CA, Simpson C, Vetto JT, Rosenberg SA. High-dose recombinant interleukin 2 in the treatment of patients with disseminated cancer. Responses, treatment-related morbidity, and histologic findings. JAMA. 1986;256(22):3117–3124. [PubMed] [Google Scholar]
  • 10.Rosenberg SA. Raising the bar: the curative potential of human cancer immunotherapy. Sci. Transl. Med. 2012;4(127):S128. doi: 10.1126/scitranslmed.3003634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Malek TR, Castro I. Interleukin-2 receptor signaling: at the interface between tolerance and immunity. Immunity. 2010;33(2):153–165. doi: 10.1016/j.immuni.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Liao W, Lin JX, Leonard WJ. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity. 2013;38(1):13–25. doi: 10.1016/j.immuni.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Siegel JP, Sharon M, Smith PL, Leonard WJ. The IL-2 receptor beta chain (p70): role in mediating signals for LAK, NK, and proliferative activities. Science. 1987;238(4823):75–78. doi: 10.1126/science.3116668. [DOI] [PubMed] [Google Scholar]
  • 14.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4 + CD25 + regulatory T-cells. Nat. Immunol. 2003;4(4):330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 15.Saadoun D, Rosenzwajg M, Joly F, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N. Engl. J. Med. 2011;365(22):2067–2077. doi: 10.1056/NEJMoa1105143. [DOI] [PubMed] [Google Scholar]
  • 16.Lee PP, Yee C, Savage PA, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat. Med. 1999;5(6):677–685. doi: 10.1038/9525. [DOI] [PubMed] [Google Scholar]
  • 17.Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat. Med. 1998;4(3):321–327. doi: 10.1038/nm0398-321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Dudley ME, Wunderlich JR, Robbins PF, et al. Cancer regression and autoimmuniky in patients after clonal repopulation with antitkumor lymphocytes. Science. 2002;298(5594):850–854. doi: 10.1126/science.1076514. • First clinical demonstration that lymphodepletion prior to adoptive immunotherapy enhances transferred T-cell persistence and elicits melanoma tumor regression.
  • 19. Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. Selective stimulation of T-cell subsets with antibody-cytokine immune complexes. Science. 2006;311(5769):1924–1927. doi: 10.1126/science.1122927. • First demonstration of how IL-2/anti-IL-2 complexes can differentially target CD8+ T cells and Tregs in vivo.
  • 20.Fishman MN, Thompson JA, Pennock GK, et al. Phase I trial of ALT-801, an interleukin-2/T-cell receptor fusion protein targeting p53 (aa264–272)/HLA-A*0201 complex, in patients with advanced malignancies. Clin. Cancer Res. 2011;17(24):7765–7775. doi: 10.1158/1078-0432.CCR-11-1817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Levin AM, Bates DL, Ring AM, et al. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature. 2012;484(7395):529–533. doi: 10.1038/nature10975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Rochman Y, Spolski R, Leonard WJ. New insights into the regulation of T cells by gamma(c) family cytokines. Nat.Rev. Immunol. 2009;9(7):480–490. doi: 10.1038/nri2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Teague RM, Sather BD, Sacks JA, et al. Interleukin-15 rescues tolerant CD8+ T cells for use in adoptive immunotherapy of established tumors. Nat. Med. 2006;12(3):335–341. doi: 10.1038/nm1359. [DOI] [PubMed] [Google Scholar]
  • 24.Hinrichs CS, Spolski R, Paulos CM, et al. IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy. Blood. 2008;111(11):5326–5333. doi: 10.1182/blood-2007-09-113050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Waldmann TA. The biology of interleukin-2 and interleukin-15: implications for cancer therapy and vaccine design. Nat. Rev. Immunol. 2006;6(8):595–601. doi: 10.1038/nri1901. [DOI] [PubMed] [Google Scholar]
  • 26.Rubinstein MP, Kovar M, Purton JF, et al. Converting IL-15 to a superagonist by binding to soluble IL-15R{alpha} Proc. Natl Acad. Sci. USA. 2006;103(24):9166–9171. doi: 10.1073/pnas.0600240103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Han KP, Zhu X, Liu B, et al. IL-15:IL-15 receptor alpha superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization. Cytokine. 2011;56(3):804–810. doi: 10.1016/j.cyto.2011.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Xu W, Jones M, Liu B, et al. Efficacy and mechanism-of-action of a novel superagonist interleukin-15: interleukin-15 receptor alphaSu/Fc fusion complex in syngeneic murine models of multiple myeloma. Cancer Res. 2013;73(10):3075–3086. doi: 10.1158/0008-5472.CAN-12-2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Spolski R, Leonard WJ. Interleukin-21: basic biology and implications for cancer and autoimmuniky. Annu. Rev. Immunol. 2008;26:57–79. doi: 10.1146/annurev.immunol.26.021607.090316. [DOI] [PubMed] [Google Scholar]
  • 30.Zeng R, Spolski R, Finkelstein SE, et al. Synergy of IL-21 and IL-15 in regulating CD8+ T-cell expansion and function. J. Exp. Med. 2005;201(1):139–148. doi: 10.1084/jem.20041057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Clough LE, Wang CJ, Schmidt EM, et al. Release from regulatory T-cell-mediated suppression during the onset of tissue-specific autoimmuniky is associated with elevated IL-21. J. Immunol. 2008;180(8):5393–5401. doi: 10.4049/jimmunol.180.8.5393. [DOI] [PubMed] [Google Scholar]
  • 32.Li Y, Yee C. IL-21 mediated Foxp3 suppression leads to enhanced generation of antigen-specific CD8+ cytotoxic T lymphocytes. Blood. 2008;111(1):229–235. doi: 10.1182/blood-2007-05-089375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Peluso I, Fantini MC, Fina D, et al. IL-21 counteracts the regulatory T-cell-mediated suppression of human CD4 + T lymphocytes. J. Immunol. 2007;178(2):732–739. doi: 10.4049/jimmunol.178.2.732. [DOI] [PubMed] [Google Scholar]
  • 34.Jung G, Ledbetter JA, Muller-Eberhard HJ. Induction of cytotoxicity in resting human T lymphocytes bound to tumor cells by antibody heteroconjugates. Proc. Natl Acad. Sci. USA. 1987;84(13):4611–4615. doi: 10.1073/pnas.84.13.4611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 2006;355(10):1018–1028. doi: 10.1056/NEJMoa063842. [DOI] [PubMed] [Google Scholar]
  • 36.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]
  • 37.Stromnes IM, Schmitt TM, Chapuis AG, Hingorani SR, Greenberg PD. Re-adapting T cells for cancer therapy: from mouse models to clinical trials. Immunol. Rev. 2014;257(1):145–164. doi: 10.1111/imr.12141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cheadle EJ, Gornall H, Baldan V, Hanson V, Hawkins RE, Gilham DE. CAR T-cells: driving the road from the laboratory to the clinic. Immunol. Rev. 2014;257(1):91–106. doi: 10.1111/imr.12126. [DOI] [PubMed] [Google Scholar]
  • 39.Mallett S, Fossum S, Barclay AN. Characterization of the MRC OX40 antigen of activated CD4 positive T lymphocytes-a molecule related to nerve growth factor receptor. Embo. J. 1990;9(4):1063–1068. doi: 10.1002/j.1460-2075.1990.tb08211.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Godfrey WR, Fagnoni FF, Harara MA, Buck D, Engleman EG. Identification of a human OX-40 ligand, a costimulator of CD4+ T cells with homology to tumor necrosis factor. J. Exp. Med. 1994;180(2):757–762. doi: 10.1084/jem.180.2.757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Watts TH. TNF/TNFR family members in costimulation of T-cell responses. Annu. Rev. Immunol. 2005;23:23–68. doi: 10.1146/annurev.immunol.23.021704.115839. [DOI] [PubMed] [Google Scholar]
  • 42.Gramaglia I, Weinberg AD, Lemon M, Croft M. Ox-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T-cell responses. J. Immunol. 1998;161(12):6510–6517. [PubMed] [Google Scholar]
  • 43.Rogers PR, Song J, Gramaglia I, Killeen N, 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):445–455. doi: 10.1016/s1074-7613(01)00191-1. [DOI] [PubMed] [Google Scholar]
  • 44.Gough MJ, Ruby CE, Redmond WL, Dhungel B, Brown A, Weinberg AD. OX40 agonist therapy enhances CD8 infiltration and decreases immune suppression in the tumor. Cancer Res. 2008;68(13):5206–5215. doi: 10.1158/0008-5472.CAN-07-6484. [DOI] [PubMed] [Google Scholar]
  • 45.Vu MD, Xiao X, Gao W, et al. OX40 costimulation turns off Foxp3+ Tregs. Blood. 2007;110(7):2501–2510. doi: 10.1182/blood-2007-01-070748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Vetto JT, Lum S, Morris A, et al. Presence of the T-cell activation marker OX-40 on tumor infiltrating lymphocytes and draining lymph node cells from patients with melanoma and head and neck cancers. Am. J. Surg. 1997;174(3):258–265. doi: 10.1016/s0002-9610(97)00139-6. [DOI] [PubMed] [Google Scholar]
  • 47.Curti BD, Kovacsovics-Bankowski M, Morris N, et al. 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]
  • 48.Hernandez-Chacon JA, Li Y, Wu RC, et al. Costimulation through the CD137/4—IBB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitkumor effector function. J. Immunother. 2011;34(3):236–250. doi: 10.1097/CJI.0b013e318209e7ec. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Melero I, Shuford WW, Newby SA, et al. Monoclonal antibodies against the 4-IBB T-cell activation molecule eradicate established tumors. Nat. Med. 1997;3(6):682–685. doi: 10.1038/nm0697-682. [DOI] [PubMed] [Google Scholar]
  • 50.Shuford WW, Klussman K, Tritchler DD, et al. 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]
  • 51.Wilcox RA, Flies DB, Zhu G, et al. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J. Clin. Invest. 2002;109(5):651–659. doi: 10.1172/JCI14184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Molckovsky A, Ski LL. First-in-class, first-in-human Phase I results of targeted agents: highlights of the 2008 American society of clinical oncology meeting. J. Hematol. Oncol. 2008;1:20. doi: 10.1186/1756-8722-1-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sotomayor EM, Borrello I, Tubb E, et al. Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40. Nat. Med. 1999;5(7):780–787. doi: 10.1038/10503. [DOI] [PubMed] [Google Scholar]
  • 54.Diehl L, Den Boer AT, Schoenberger SP, et al. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments antitkumor vaccine efficacy. Nat. Med. 1999;5(7):774–779. doi: 10.1038/10495. [DOI] [PubMed] [Google Scholar]
  • 55.French RR, Chan HT, Tutt AL, Glennie MJ. CD40 antibody evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help. Nat. Med. 1999;5(5):548–553. doi: 10.1038/8426. [DOI] [PubMed] [Google Scholar]
  • 56.Zhang L, Chen X, Liu X, et al. CD40 ligation reverses T-cell tolerance in acute myeloid leukemia. J. Clin. Invest. 2013;123(5):1999–2010. doi: 10.1172/JCI63980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bensinger W, Maziarz RT, Jagannath S, et al. A Phase 1 study of lucatumumab, a fully human anti-CD40 antagonist monoclonal antibody administered intravenously to patients with relapsed or refractory multiple myeloma. Br. J. Haematol. 2012;159(1):58–66. doi: 10.1111/j.1365-2141.2012.09251.x. [DOI] [PubMed] [Google Scholar]
  • 58.Byrd JC, Kipps TJ, Flinn IW, et al. Phase I study of the anti-CD40 humanized monoclonal antibody lucatumumab (HCD122) in relapsed chronic lymphocytic leukemia. Leuk. Lymphoma. 2012;53(11):2136–2142. doi: 10.3109/10428194.2012.681655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Furman RR, Forero-Torres A, Shustov A, Drachman JG. A Phase I study of dacetuzumab (SGN-40, a humanized anti-CD40 monoclonal antibody) in patients with chronic lymphocytic leukemia. Leuk. Lymphoma. 2010;51(2):228–235. doi: 10.3109/10428190903440946. [DOI] [PubMed] [Google Scholar]
  • 60.Hussein M, Berenson JR, Niesvizky R, et al. A Phase I multidose study of dacetuzumab (SGN-40; humanized anti-CD40 monoclonal antibody) in patients with multiple myeloma. Haematologica. 2010;95(5):845–848. doi: 10.3324/haematol.2009.008003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vonderheide RH, Flaherty KT, Khalil M, et al. Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J. Clin. Oncol. 2007;25(7):876–883. doi: 10.1200/JCO.2006.08.3311. [DOI] [PubMed] [Google Scholar]
  • 62.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 63.Hawiger D, Inaba K, Dorsett Y, et al. Dendritic cells induce peripheral T-cell unresponsiveness under steady state conditions in vivo . J. Exp. Med. 2001;194(6):769–779. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Banchereau J, Thompson-Snipes L, Zurawski S, et al. The differential production of cytokines by human Langerhans cells and dermal CD14(+) DCs controls CTL priming. Blood. 2012;119(24):5742–5749. doi: 10.1182/blood-2011-08-371245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chen L, Ashe S, Brady WA, et al. Costimulation of antitkumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell. 1992;71(7):1093–1102. doi: 10.1016/s0092-8674(05)80059-5. [DOI] [PubMed] [Google Scholar]
  • 66.Ueno H, Schmkt N, Klechevsky E, et al. Harnessing human dendritic cell subsets for medicine. Immunol. Rev. 2010;234(1):199–212. doi: 10.1111/j.0105-2896.2009.00884.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat. Rev. Cancer. 2012;12(4):265–277. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010;363(5):411–422. doi: 10.1056/NEJMoa1001294. • Clinical report showing personalized commercial DC vaccine (Sipuleucel-T) prolongs survival of patients with prostate cancer.
  • 69.Bonifaz LC, Bonnyay DP, Charalambous A, et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T-cell vaccination. J. Exp. Med. 2004;199(6):815–824. doi: 10.1084/jem.20032220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ribas A, Comin-Anduix B, Chmielowski B, et al. Dendritic cell vaccination combined with CTLA4 blockade in patients with metastatic melanoma. Clin. Cancer Res. 2009;15(19):6267–6276. doi: 10.1158/1078-0432.CCR-09-1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bahjat KS, Liu W, Lemmens EE, et al. Cytosolic entry controls CD8+ -T-cell potency during bacterial infection. Infect. Immun. 2006;74(11):6387–6397. doi: 10.1128/IAI.01088-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Souders NC, Sewell DA, Pan ZK, et al. Listeria-based vaccines can overcome tolerance by expanding low avidity CD8+ T cells capable of eradicating a solid tumor in a transgenic mouse model of cancer. Cancer Immun. 2007;7:2. [PMC free article] [PubMed] [Google Scholar]
  • 73.Na SY, Hermann A, Sanchez-Ruiz M, Storch A, Deckert M, Hunig T. Oligodendrocytes enforce immune tolerance of the uninfected brain by purging the peripheral repertoire of autoreactive CD8+ T cells. Immunity. 2012;37(1):134–146. doi: 10.1016/j.immuni.2012.04.009. [DOI] [PubMed] [Google Scholar]
  • 74.Shahabi V, Reyes-Reyes M, Wallecha A, Rivera S, Paterson Y, Maciag P. Development of a Listeria monocytogenes based vaccine against prostate cancer. Cancer Immunol. Immunother. 2008;57(9):1301–1313. doi: 10.1007/s00262-008-0463-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Gunn GR, Zubair A, Peters C, Pan ZK, Wu TC, Paterson Y. Two Listeria monocytogenes vaccine vectors that express different molecular forms of human papilloma virus-16 (HPV-16) E7 induce qualitatively different T-cell immunity that correlates with their ability to induce regression of established tumors immortalized by HPV-16. J. Immunol. 2001;167(11):6471–6479. doi: 10.4049/jimmunol.167.11.6471. [DOI] [PubMed] [Google Scholar]
  • 76.Maciag PC, Radulovic S, Rothman J. The first clinical use of a live-attenuated Listeria monocytogenes vaccine: a Phase I safety study of Lm-LLO-E7 in patients with advanced carcinoma of the cervix. Vaccine. 2009;27(30):3975–3983. doi: 10.1016/j.vaccine.2009.04.041. [DOI] [PubMed] [Google Scholar]
  • 77.Le DT, Brockstedt DG, Nir-Paz R, et al. A live-attenuated Listeria vaccine (ANZ-100) and a live-attenuated Listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: phase I studies of safety and immune induction. Clin. Cancer Res. 2012;18(3):858–868. doi: 10.1158/1078-0432.CCR-11-2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat. Rev. Cancer. 2008;8(4):299–308. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Schmitt TM, Ragnarsson GB, Greenberg PD. T-cell receptor gene therapy for cancer. Hum. Gene Ther. 2009;20(11):1240–1248. doi: 10.1089/hum.2009.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314(5796):126–129. doi: 10.1126/science.1129003. • First clinical use of genetically engineered T-cell receptor for adoptive T-cell immunotherapy in patients with melanoma.
  • 81.Schmitt TM, Aggen DH, Stromnes IM, et al. Enhanced-affinity murine T-cell receptors for tumor/self-antigens can be safe in gene therapy despite surpassing the threshold for thymic selection. Blood. 2013;122(3):348–356. doi: 10.1182/blood-2013-01-478164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Robbins PF, Morgan RA, Feldman SA, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J. Clin. Oncol. 2011;29(7):917–924. doi: 10.1200/JCO.2010.32.2537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Morgan RA, Chinnasamy N, Abate-Daga D, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 2013;36(2):133–151. doi: 10.1097/CJI.0b013e3182829903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Linette GP, Stadtmauer EA, Maus MV, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122(6):863–871. doi: 10.1182/blood-2013-03-490565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T-cell response. Nat. Rev. Immunol. 2012;12(4):269–281. doi: 10.1038/nri3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Song DG, Ye Q, Carpenito C, et al. In vivo persistence, tumor localization, and antitkumor activity of CAR-engineered T-cells is enhanced by costimulatory signaling through CD137 (4–1BB) Cancer Res. 2011;71(13):4617–4627. doi: 10.1158/0008-5472.CAN-11-0422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013;368(16):1509–1518. doi: 10.1056/NEJMoa1215134. • Highlights the use of engineered chimeric antigen receptor T-cells for treatment of patients with leukemia.
  • 88.Sadelain M, Brentjens R, Riviere I. The promise and potential pitfalls of chimeric antigen receptors. Curr. Opin. Immunol. 2009;21(2):215–223. doi: 10.1016/j.coi.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.ClinicalTrials.Gov. www.ClinicalTrials.gov.
  • 90.Chmielewski M, Hombach AA, Abken H. Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol. Rev. 2014;257(1):83–90. doi: 10.1111/imr.12125. [DOI] [PubMed] [Google Scholar]
  • 91.Chmielewski M, Kopecky C, Hombach AA, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011;71(17):5697–5706. doi: 10.1158/0008-5472.CAN-11-0103. [DOI] [PubMed] [Google Scholar]
  • 92.Pegram HJ, Lee JC, Hayman EG, et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012;119(18):4133–4141. doi: 10.1182/blood-2011-12-400044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chmielewski M, Abken H. CAR T cells transform to trucks: chimeric antigen receptor-redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer. Cancer Immunol. Immunother. 2012;61(8):1269–1277. doi: 10.1007/s00262-012-1202-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, 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]
  • 95.Brentjens R, Yeh R, Bernal Y, Riviere I, Sadelain M. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a Phase I clinical trial. Mol. Ther. 2010;18(4):666–668. doi: 10.1038/mt.2010.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Budde LE, Berger C, Lin Y, et al. 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]
  • 97.Goldrath AW, Bogatzki LY, Bevan MJ. Naive T cells transiently acquire a memory-like phenotype during homeostasis-driven proliferation. J. Exp. Med. 2000;192(4):557–564. doi: 10.1084/jem.192.4.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cho BK, Rao VP, Ge Q, Eisen HN, Chen J. Homeostasis-stimulated proliferation drives naive T cells to differentiate directly into memory T cells. J. Exp. Med. 2000;192(4):549–556. doi: 10.1084/jem.192.4.549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Schietinger A, Delrow JJ, Basom RS, Blattman JN, Greenberg PD. Rescued tolerant CD8 T cells are preprogrammed to reestablish the tolerant state. Science. 2012;335(6069):723–727. doi: 10.1126/science.1214277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Eberlein TJ, Rosenstein M, Rosenberg SA. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J. Exp. Med. 1982;156(2):385–397. doi: 10.1084/jem.156.2.385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.North RJ. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T-cells. J. Exp. Med. 1982;155(4):1063–1074. doi: 10.1084/jem.155.4.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J. Clin. Oncol. 2005;23(10):2346–2357. doi: 10.1200/JCO.2005.00.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 2005;202(7):907–912. doi: 10.1084/jem.20050732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011;17(13):4550–4557. doi: 10.1158/1078-0432.CCR-11-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004;10(9):942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 106.Schubert LA, Jeffery E, Zhang Y, Ramsdell F, Ziegler SF. Scurfin (FOXP3) acts as a repressor of transcription and regulates T-cell activation. J. Biol. Chem. 2001;276(40):37672–37679. doi: 10.1074/jbc.M104521200. [DOI] [PubMed] [Google Scholar]
  • 107.Savage PA, Malchow S, Leventhal DS. Basic principles of tumor-associated regulatory T-cell biology. Trends Immunol. 2013;34(1):33–40. doi: 10.1016/j.it.2012.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Dannull J, Su Z, Rizzieri D, et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J. Clin. Invest. 2005;115(12):3623–3633. doi: 10.1172/JCI25947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Rech AJ, Vonderheide RH. Clinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cells. Ann. NY Acad. Sci. 2009;1174:99–106. doi: 10.1111/j.1749-6632.2009.04939.x. [DOI] [PubMed] [Google Scholar]
  • 110.Jacobs JF, Punt CJ, Lesterhuis WJ, et al. Dendritic cell vaccination in combination with anti-CD25 monoclonal antibody treatment: a Phase I/II study in metastatic melanoma patients. Clin. Cancer Res. 2010;16(20):50675078. doi: 10.1158/1078-0432.CCR-10-1757. [DOI] [PubMed] [Google Scholar]
  • 111. Hodi FS, Butler M, Oble DA, et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc. Natl Acad. Sci. USA. 2008;105(8):3005–3010. doi: 10.1073/pnas.0712237105. • One of the first clinical studies showing the efficacy of anti-CTLA-4 checkpoint blockade in patients with melanoma.
  • 112.Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 2013 doi: 10.1084/jem.20130579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Almand B, Clark JI, Nikitina E, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J. Immunol. 2001;166(1):678–689. doi: 10.4049/jimmunol.166.1.678. [DOI] [PubMed] [Google Scholar]
  • 114.Ostrand-Rosenberg S. Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol. Immunother. 2010;59(10):1593–1600. doi: 10.1007/s00262-010-0855-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI. Mechanism of T-cell tolerance induced by myeloid-derived suppressor cells. J. Immunol. 2010;184(6):3106–3116. doi: 10.4049/jimmunol.0902661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol. Immunother. 2009;58(1):49–59. doi: 10.1007/s00262-008-0523-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Najjar YG, Finke JH. Clinical perspectives on targeting of myeloid derived suppressor cells in the treatment of cancer. Front. Oncol. 2013;3:49. doi: 10.3389/fonc.2013.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002;3(7):611–618. doi: 10.1038/ni0702-611. [DOI] [PubMed] [Google Scholar]
  • 119.Waterhouse P, Penninger JM, Timms E, et al. 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]
  • 120.Lee KM, Chuang E, Griffin M, et al. Molecular basis of T-cell inactivation by CTLA-4. Science. 1998;282(5397):2263–2266. doi: 10.1126/science.282.5397.2263. [DOI] [PubMed] [Google Scholar]
  • 121.Guntermann C, Alexander DR. CTLA-4 suppresses proximal TCR signaling in resting human CD4 (+) T cells by inhibiting ZAP-70 Tyr(319) phosphorylation: a potential role for tyrosine phosphatases. J. Immunol. 2002;168(9):4420–4429. doi: 10.4049/jimmunol.168.9.4420. [DOI] [PubMed] [Google Scholar]
  • 122.Qureshi OS, Zheng Y, Nakamura K, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332(6029):600–603. doi: 10.1126/science.1202947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hodi FS, O’day SJ, Mcdermott DF, et al. 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]
  • 124.Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal infammation. J. Exp. Med. 2000;192(2):295–302. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J. Exp. Med. 2000;192(2):303–310. doi: 10.1084/jem.192.2.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat. Immunol. 2007;8(3):239–245. doi: 10.1038/ni1443. [DOI] [PubMed] [Google Scholar]
  • 127.Barber DL, Wherry EJ, Masopust D, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439(7077):682–687. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
  • 128.Berrien-Elliott MM, Jackson SR, Meyer JM, et al. Durable adoptive immunotherapy for leukemia produced by manipulation of multiple regulatory pathways of CD8+ T-cell tolerance. Cancer Res. 2013;73(2):605–616. doi: 10.1158/0008-5472.CAN-12-2179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114(8):1545–1552. doi: 10.1182/blood-2009-03-206672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T-cell proliferation. Sci. Signal. 2012;5(230):ra46. doi: 10.1126/scisignal.2002796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat. Med. 2002;8(8):793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 132.Brahmer JR, Tykodi SS, Chow LQ, et al. 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]
  • 133.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012;366(26):2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 2013;369(2):134–144. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Weber JS, Kahler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J. Clin. Oncol. 2012;30(21):2691–2697. doi: 10.1200/JCO.2012.41.6750. [DOI] [PubMed] [Google Scholar]
  • 136.Gangadhar TC, Vonderheide RH. Mitigating the toxic effects of anticancer immunotherapy. Nat. Rev. Clin. Oncol. 2014;11(2):91–99. doi: 10.1038/nrclinonc.2013.245. [DOI] [PubMed] [Google Scholar]
  • 137.Huang CT, Workman CJ, Flies D, et al. 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]
  • 138.Sabatos CA, Chakravarti S, Cha E, et al. 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]
  • 139.Sanchez-Fueyo A, Tian J, Picarella D, et al. Tim-3 inhibits T helper type 1-mediated auto- and alloimmune responses and promotes immunological tolerance. Nat. Immunol. 2003;4(11):1093–1101. doi: 10.1038/ni987. [DOI] [PubMed] [Google Scholar]
  • 140.Teague RM, Kline J. Immune evasion in acute myeloid leukemia: current concepts and future directions. J. Immunother. Cancer. 2013;1(13) doi: 10.1186/2051-1426-1-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Riley JL. Combination checkpoint blockade-taking melanoma immunotherapy to the next level. N. Engl. J. Med. 2013;369(2):187–189. doi: 10.1056/NEJMe1305484. [DOI] [PubMed] [Google Scholar]
  • 142.Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. USA. 2010;107(9):4275–4280. doi: 10.1073/pnas.0915174107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Grosso JF, Goldberg MV, Getnet D, et al. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J. Immunol. 2009;182(11):6659–6669. doi: 10.4049/jimmunol.0804211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T-cell exhaustion and restore antitumor immunity. J. Exp. Med. 2010;207(10):2187–2194. doi: 10.1084/jem.20100643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 2013;369(2):122–133. doi: 10.1056/NEJMoa1302369. • First clinical study of combination checkpoint blockade immunotherapy showing synergistic antitumor effects with a manageable safety profile.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementrary

RESOURCES