Abstract
Immunostimulatory monoclonal antibodies (mAbs) exert antineoplastic effects by eliciting a novel or reinstating a pre-existing antitumor immune response. Most often, immunostimulatory mAbs activate T lymphocytes or natural killer (NK) cells by inhibiting immunosuppressive receptors, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1 (PDCD1, best known as PD-1), or by engaging co-stimulatory receptors, like CD40, tumor necrosis factor receptor superfamily, member 4 (TNFRSF4, best known as OX40) or TNFRSF18 (best known as GITR). The CTLA4-targeting mAb ipilimumab has been approved by the US Food and Drug Administration for use in patients with unresectable or metastatic melanoma in 2011. The therapeutic profile of ipilimumab other CTLA4-blocking mAbs, such as tremelimumab, is currently being assessed in subjects affected by a large panel of solid neoplasms. In the last few years, promising clinical results have also been obtained with nivolumab, a PD-1-targeting mAb formerly known as BMS-936558. Accordingly, the safety and efficacy of nivolumab and other PD-1-blocking molecules are being actively investigated. Finally, various clinical trials are underway to test the therapeutic potential of OX40- and GITR-activating mAbs. Here, we summarize recent findings on the therapeutic profile of immunostimulatory mAbs and discuss clinical trials that have been launched in the last 14 months to assess the therapeutic profile of these immunotherapeutic agents.
Keywords: CD137, checkpoint blockade, immunogenic chemotherapy, immunosuppression, lirilumab, IPH2101, PD-L1
Introduction
A large panel of monoclonal antibodies (mAbs) is currently approved by the US Food and Drug Administration (FDA) and other international regulatory agencies, including the European Medicines Agency (EMA), for the treatment of conditions as diverse as autoimmune diseases and cancer.1,2 For illustrative purposes, antineoplastic mAbs can be subdivided into 2 large groups: (1) tumor-targeting mAbs, which directly bind to malignant cells or intercept trophic signals delivered by the tumor stroma;2 and (2) immunostimulatory mAbs, which operate by interacting with (hence modulating the function of) components of the immune system.3-5
As we have discussed in previous issues of OncoImmunology,6-8 the therapeutic potential of tumor-targeting mAbs may or may not involve immune effectors. Thus, while some of these molecules, such as the vascular endothelial growth factor (VEGF)-specific IgG1 bevacizumab,9,10 mainly exert antineoplastic effects by inhibiting pro-survival or mitogenic signaling pathways, others, such as the CD20-targeting IgG1 rituximab,11-13 near-to-completely rely on effector mechanisms of innate immunity, including antibody-dependent cell-mediated cytotoxicity (ADCC),2,14-17 antibody-dependent cellular phagocytosis (ADCP),18 and complement-dependent cytotoxicity (CDC).19,20 Of note, some tumor-targeting mAbs, such as cetuximab, a chimeric IgG1 specific for the epidermal growth factor receptor (EGFR),21,22 appear to inhibit tumor growth via both cancer cell-autonomous and immune system-dependent mechanisms.23-26 In addition, tumor-targeting mAbs can be harnessed as carriers for the selective delivery to malignant cells of toxins or radionuclides. This is the case of the CD20-targeting molecules 90Y-ibritumomab tiuxetan and 131I-tositumomab, which are currently approved for the treatment of non-Hodgkin’s lymphoma (NHL).27,28
The efficacy of immunostimulatory mAbs invariably relies on the elicitation of a novel or on the reactivation of a pre-existing immune response against malignant cells.3-5 So far, this has been achieved through three general strategies: (1) the blockade of inhibitory receptors such as cytotoxic T lymphocyte-associated protein 4 (CTLA4)29-31 and programmed cell death 1 (PDCD1, best known as PD-1),32-36 both of which are expressed by activated T lymphocytes, or various members of the killer cell immunoglobulin-like receptor (KIR) family, which are found on the surface of natural killer (NK) cells;37-40 (2) the activation of co-stimulatory receptors such as tumor necrosis factor receptor superfamily, member 4 (TNFRSF4, best known as OX40)41-44 and TNFRSF18 (best known as GITR),45,46 which are expressed by activated CD4+ and CD8+ T cells; (3) the neutralization of soluble immunosuppressive factors, such as transforming growth factor β1 (TGFβ1) (Table 1).47-51
Table 1. Immunostimulatory mAbs currently in clinical development.
Target(s) | mAb | Aliases | Isotype | Source | Activity | Owner |
---|---|---|---|---|---|---|
CD27 | CDX-1127 | 1F5 | n.a. | Human | Agonist | Celldex |
CD40 | Chi Lob 7/4 | - | IgG1 | Chimeric | Agonist | Cancer Research UK |
CP-870,893 | - | IgG2 | Human | Agonist | Pfizer | |
Dacetuzumab | SGN-40 huS2C6 |
IgG1 | Humanized | Agonist | Seattle Genetics | |
Lucatumumab | HCD122 | IgG1 | Human | Agonist | Novartis Pharmaceuticals | |
CD274 (PD-L1, B7-H1) |
BMS-936559 | MDX-1105 | IgG4 | Human | Antagonist | Bristol–Myers Squibb |
MEDI4736 | - | IgG1 | Human | Antagonist | Medimmune | |
MPDL3280A | RG7446 | IgG1 | Human | Antagonist | Genentech (Roche) | |
MSB0010718C | - | n.a. | Human | Antagonist | Serono (Merck) | |
CTLA4 | Ipilimumab | BMS-734016 MDX-010 MDX-101 Yervoy® |
IgG1κ | Human | Antagonist | Bristol–Myers Squibb |
Tremelimumab | CP-675,206 Ticilimumab |
IgG2 | Human | Antagonist | Pfizer | |
KIR | IPH2101 | 1–7F9 | IgG4 | Human | Antagonist | Innate Pharma |
Lirilumab | BMS-986015 IPH2102 |
IgG4 | Human | Antagonist | Bristol–Myers Squibb | |
PDCD1 (PD-1) |
AMP-224 | - | IgG1 chimera | Human | Antagonist | Amplimmune (Medimmune, AstraZeneca) |
Lambrolizumab | MK-3475 | IgG4 | Humanized | Antagonist | Merck | |
Nivolumab | BMS-936558 MDX1106 ONO-4538 | IgG4 | Human | Antagonist | Bristol–Myers Squibb | |
Pidilizumab | CT-011 | IgG1κ | Humanized | Antagonist | CureTech | |
MHCII | IMP321 | - | IgG1 chimera | Human | Agonist | Immutep |
PDCD1LG2 (PD-L2, B7-DC) |
rHIgM12B7 | - | IgM | Human | Agonist | Mayo Foundation |
TGFβ1 | Fresolimumab | GC1008 | IgG4κ | Human | Antagonist | Sanofi–Aventis |
TNFRSF4 (OX40) |
9B12 | - | IgG1 | Murine | Agonist | AgonOx |
MEDI6469 | - | n.a. | Murine | Agonist | Medimmune (AstraZeneca) |
|
TNFRSF9 (CD137, 4–1BB) |
Urelumab | BMS-663513 | IgG4 | Human | Agonist | Bristol-Myers Squibb |
PF-05082566 | - | IgG2 | Human | Agonist | Pfizer | |
TNFRSF18 (GITR) |
TRX518 | - | IgG1 | Humanized | Agonist | Tolerex |
Abbreviations: CTLA4, cytotoxic T lymphocyte-associated protein 4; KIR, killer cell immunoglobulin-like receptor; mAb, monoclonal antibody; MHCII, MHC Class II; n.a., not available; PDCD1, programmed cell death 1; PDCD1LG2, PDCD1 ligand 2; TGFβ1, transforming growth factor β1; TNFRSF, tumor necrosis factor receptor superfamily.
At odds with their tumor-targeting counterparts, which have attracted attention as potential anticancer agents as early as in the 1980s,52-55 immunostimulatory mAbs have been the focus of intensive preclinical and clinical investigation only with the advent of the 21st century. At least in part, this relates to the fact that tumors have long been considered as immunologically silent entities, a notion that begun to change only in the late 1990s, thanks to the theoretical foundations provided by Polly Matzinger’s “danger theory.”56,57 In spite of such a delayed kickoff, however, the clinical development of immunostimulatory antibodies has proceeded at a rapid pace, culminating in 2011 with the approval of ipilimumab, a fully human CTLA4-targeting IgG1κ also known as MDX-010, MDX-101, and BMS-734016 (now commercialized by Bristol–Myers Squibb under the trade mark Yervoy®), for use in patients with unresectable or metastatic melanoma.58-60 Ipilimumab nowadays represents the sole immunostimulatory mAb licensed by regulatory agencies for use in cancer patients, whereas no less than 14 tumor-targeting mAbs are currently employed in the clinic as part of FDA-approved immunotherapeutic regimens.1,2,8 This said, starting with the late 2000s, promising results have also been obtained in clinical trials investigating the safety and therapeutic profile of other immunostimulatory mAbs, including (1) the PD-1-targeting molecules nivolumab, a human IgG4 also known as BMS-936558, MDX-1106, and ONO-4538,61-64 and lambrolizumab, a humanized IgG4 also known as MK-3475;65 (2) BMS-936559 (also known as MDX-1105), a human IgG4 that targets the PD-1 ligand CD274 (best known as PD-L1);66 and (3) the CD40 agonist mAbs CP-870,893 (a human IgG2),67 and dacetuzumab, a humanized IgG1 also known as SGN-40 and huS2C6.68,69
Fully reflecting their immunostimulatory nature, mAbs may provoke immune reactions against self antigens that, at least potentially, may result in a life-threatening functional and/or structural damage to healthy tissues.3,4 However, these reactions most often fail to reach a clinically meaningful amplitude and can be controlled with corticosteroids or other immunosuppressants.3,4 Thus, immunostimulatory mAbs stand out as a relatively safe and well tolerated immunotherapeutic regimen, most frequently causing mild and controllable adverse effects including (but not limited to) skin rashes, pruritus, fatigue, nausea, and diarrhea.3,4 As a standalone exception, urelumab (also known as BMS-663513), which operates as an agonist of the co-stimulatory receptor TNFRSF9 (best known as CD137 or 4–1BB), has been associated with severe hepatotoxicity (in particular when employed at high doses).70,71
We have summarized recent advances on the clinical use of tumor-targeting mAbs in the latest issue of OncoImmunology.8 Here, along the lines of our monthly Trial Watch series,72-75 we will restrict our focus on immunostimulatory antibodies, discussing the results of clinical trials published in the last 14 mo and commenting on studies launched in the same period to evaluate the safety and efficacy of this immunotherapeutic paradigm in cancer patients.
Update on Clinical Reports
During the last 14 mo, the results of no less than 19 clinical trials investigating the therapeutic potential of immunostimulatory mAbs in cancer patients have been published in peer-reviewed scientific journals (source http://www.ncbi.nlm.nih.gov/pubmed). More than half of these studies involved mAbs that antagonize the delivery of inhibitory signals to immune effector cells, including the CTLA4-targeting agents ipilimumab76-82 and tremelimumab (a human IgG2 also known as ticilimumab or CP-675,206),83-85 the PD-1-specific mAbs nivolumab and lambrolizumab,82,86,87 as well as IPH2101 (a KIR-inhibitory human IgG4 also known as 1-7F9).88,89 In addition, 6 studies involved mAbs that directly operate as agonists for co-stimulatory receptors, including the CD40-targeting molecules dacetuzumab, CP-870,893 and lucatumumab (a human IgG1 also known as HCD122),90-93 9B12, a murine IgG1 that triggers OX40 signaling,94 as well as IMP321, an IgG- and lymphocyte-activation gene 3 (LAG3)-based chimera that binds to, hence activating, MHC Class II molecules (Table 2).95
Table 2. Recently published clinical trials investigating the therapeutic profile of immunostimulatory mAbs.*.
Target | mAb | Indication(s) | Phase | Note | Ref. |
---|---|---|---|---|---|
CD40 | CP-870,893 | Pancreatic cancer | I | In combination with gemcitabine | 92 |
Advanced solid tumors |
I | In combination with paclitaxel and carboplatin |
90 | ||
Dacetuzumab | DLBCL | Pilot | In combination with rituximab and gemcitabine |
91 | |
Lucatumumab | CLL | I | As single agent | 93 | |
MM | I | As single agent | 108 | ||
CTLA4 | Ipilimumab | Melanoma | I | In combination with vemurafenib | 78 |
I | Combined with nivolumab | 82 | |||
III | As single agent or combined with dacarbazine |
76 | |||
Pancreatic cancer | Ib | As single agent or combined with a GM-CSF-secreting vaccine |
80 | ||
Prostate carcinoma | I/II | In combination with a GM-CSF-secreting allogeneic vaccine | 77 | ||
I/II | Combined with GM-CSF | 81 | |||
SCLC | II | In combination with paclitaxel and carboplatin |
79 | ||
Tremelimumab | Melanoma | I | As single agent | 83 | |
I | In combination with the TLR9 agonist PF-3512676 |
85 | |||
III | As single agent | 84 | |||
KIR | IPH2101 | AML | I | As single agent | 88 |
MM | I | As single agent | 89 | ||
MHCII | IMP321 | Pancreatic carcinoma | I | In combination with gemcitabine | 95 |
PDCD1 (PD-1, CD279) |
Lambrolizumab | Melanoma | I | As single agent | 87 |
Nivolumab | Melanoma | I | Combined with ipilimumab | 82 | |
Advanced solid tumors |
I | As single agent | 86 | ||
TNFRSF4 (OX40) |
9B12 | Advanced solid tumors |
I | As single agent | 94 |
Abbreviations: AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CTLA4, cytotoxic T lymphocyte-associated protein 4; DLBCL, diffuse large B-cell lymphoma; GM-CSF, granulocyte macrophage colony-stimulating factor; KIR, killer cell immunoglobulin-like receptor; mAb, monoclonal antibody; MHCII, MHC Class II; MM, multiple myeloma; PDCD1, programmed cell death 1; SCLC, small cell lung carcinoma; TLR9, Toll-like receptor 9; TNFRSF, tumor necrosis factor receptor superfamily. *between 2012, October 1st and the day of submission.
Ipilimumab has recently been tested (1) together with the BRAF inhibitor vemurafenib in patients with metastatic melanoma bearing a BRAF V600 mutation;78 (2) combined with paclitaxel (a microtubular inhibitor and hyperploidizing agent of the taxane family)96,97 and carboplatin (a second-generation platinum derivative)98,99 as first-line therapy in subjects bearing extensive small-cell lung cancer lesions;79 (3) alone or in combination with a granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting allogeneic vaccine100,101 in pancreatic ductal adenocarcinoma patients;80 and (4) as part of a nivolumab-based immunotherapeutic regimen in patients with advanced melanoma.82 In addition, (1) Sherrill and colleagues evaluated the quality-adjusted survival of patients enrolled in study CA184024, a multinational, randomized, double-blind, Phase III trial testing dacarbazine (an alkylating agent currently approved by the US FDA for the treatment of Hodgkin’s lymphoma and melanoma) alone vs. dacarbazine plus ipilimumab in subjects with previously untreated metastatic melanoma;59,76 (2) Santegoets and collaborators performed an exploratory T-cell monitoring study in the context of Phase I/II dose escalation/expansion trial testing ipilimumab plus a GM-CSF-secreting allogeneic vaccine in prostate carcinoma patients;77 and (3) Kwek and co-authors monitored the humoral immune responses elicited by the co-administration of ipilimumab and a fixed dose of GM-CSF102,103 in subjects with prostate cancer.81 In these clinical settings, ipilimumab-based immune(chemo)therapy was well tolerated and associated with promising clinical responses,79,80,82 with a single exception.78 Indeed, in 2 distinct cohorts of melanoma patients, the co-administration of ipilimumab and vemurafenib at full approved doses was associated with grade 3 elevations in circulating alanine aminotransferase (ALT) and aspartate aminotransferase (AST).78 Although such hepatotoxic effects were asymptomatic and reversible upon the temporary discontinuation of treatment or the administration of glucocorticoids, this Phase I clinical trial was promptly closed to further patient accrual.78
The clinical potential of tremelimumab, employed as a standalone therapeutic intervention or combined with the Toll-like receptor 9 (TLR9) agonist PF-3512676,104-106 has recently been assessed in patients affected by metastatic melanoma83-85 or other advanced neoplasms.85 In the context of a Phase I clinical trial, the combination of tremelimumab with PF-3512676 was well tolerated, but evoked measurable clinical responses in 2 out of 21 patients only.85 Along similar lines, in the context of a large, randomized, Phase III study, no statistically significant survival advantage could be documented in metastatic melanoma patients receiving tremelimumab vs. standard-of-care chemotherapy, although response duration (measured from the time of randomization) was significantly longer in the tremelimumab arm.84 Based on such a lack of efficacy, the A3671009 study (NCT00257205) has been discontinued.107
The safety and efficacy of PD-1-targeting mAbs have recently been investigated in 2 distinct cohorts of melanoma patients, receiving either lambrolizumab as a single agent in the context of a Phase I clinical trial (NCT01295827),87 or nivolumab coupled to ipilimumab, again as part of a Phase I study (NCT01024231).82 In addition, the long-term therapeutic potential of nivolumab given as a standalone intervention has been assessed in a mixed patient cohort, encompassing melanoma, renal cell carcinoma as well as colorectal carcinoma patients.86 In these settings, the administration of PD-1-targeting mAbs alone was associated with relatively mild (grade 1 or 2) side effects.86,87 Conversely, the co-administration of nivolumab and ipilimumab provoked grade 3 or 4 adverse reactions in a consistent proportion of melanoma patients.82 Still, (1) such side effects were qualitatively similar to those previously associated with either nivolumab or ipilimumab monotherapy and were generally reversible; and (2) the combined inhibition of CTLA4- and PD-1-dependent immunosuppression was associated with an objective response in 53% of patients, invariably manifesting with a reduction in tumor burden of 80% or more.82
The clinical potential of mAbs directly targeting NK cells, rather than T lymphocytes, has recently been assessed in patients affected by acute myeloid leukemia (EUDRACT 2005–005298–31)88 or relapsed/refractory multiple myeloma (NCT00552396).89 In both these Phase I sequential-cohort dose-escalation studies, IPH2101 was administered as a single agent, provoking limited side effects at doses that fully inhibit KIRs.88,89
Finally, the safety and efficacy of immunostimulatory mAbs that operate as agonists for co-stimulatory receptors have recently been investigated (invariably in Phase I settings) (1) in patients with chronic lymphocytic leukemia (CLL) or relapsed/refractory multiple myeloma, receiving lucatumumab as a single agent in the context of sequential-cohort dose-escalation studies;93,108 (2) in subjects affected by advanced pancreatic ductal adenocarcinoma, who were treated with CP-870,893 plus gemcitabine-based chemotherapy;92 (3) in individuals bearing advanced solid tumors, receiving CP-870,893 coupled to carboplatin and paclitaxel;90 (4) in patients with relapsed/refractory diffuse large B-cell lymphoma, who were given dacetuzumab in combination with rituximab and gemcitabine;91 (5) in individuals bearing advanced neoplasms, receiving 9B12 in combination with various adjuvants (NCT01644968);94 and (6) in patients with advanced pancreatic adenocarcinoma, receiving IMP321 plus gemcitabine-based chemotherapy.95 Globally, these immunostimulatory mAbs were well tolerated but exerted limited clinical efficacy.90-95,108
In summary, the results of the clinical studies reported above suggest that immunostimulatory mAbs are well tolerated by cancer patients, especially when administered as standalone interventions, yet elicit limited clinical responses in these conditions. Conversely, combinatorial approaches such as the co-administration of nivolumab and ipilimumab are associated not only with an increased incidence and severity of adverse effects, but also with improved clinical activity.82
Update on Clinical Trials Testing Immunostimulatory Monoclonal Antibodies
When this Trial Watch was being redacted (November 2013), official sources listed 60 clinical trials launched after 2012, October 1st to evaluate the therapeutic profile of immunostimulatory mAbs in cancer patients (source http://www.clinicaltrials.gov).
For obvious advantages related to its approval status, more than half (36) of these studies aim at investigating the clinical profile of ipilimumab, either as a standalone intervention or combined with radiotherapy, conventional chemotherapeutics, targeted anticancer drugs or other immunostimulatory agents. In particular, ipilimumab is being tested as part of immunochemotherapeutic or immunoradiotherapeutic regimens in melanoma patients (NCT01701674; NCT01703507; NCT01708941; NCT01709162; NCT01715077; NCT01730157; NCT01740297; NCT01740401; NCT01767454; NCT01769222; NCT01783938; NCT01789827; NCT01810016; NCT01827111; NCT01838200; NCT01844505; NCT01856023; NCT01866319; NCT01879306), the sole oncological indication for which this CTLA4-targeting mAb is licensed by regulatory agencies, as well as in cohorts of individuals affected by various hematological malignancies (NCT01729806; NCT01757639; NCT01769222; NCT01822509; NCT01896999; NCT01919619), prostate carcinoma (NCT01804465; NCT01832870), head and neck cancer (NCT01860430; NCT01935921), non-small cell lung carcinoma (NSCLC) (NCT01820754), cervical carcinoma (NCT01711515), Merkel cell carcinoma (NCT01913691), pancreatic cancer (NCT01896869), colorectal carcinoma (NCT01769222), and various advanced/metastatic solid tumors (NCT01738139; NCT01750580; NCT01750983; NCT01928394). Alongside, the safety and efficacy of the other CTLA4-targeting mAb tremelimumab are being assessed in cohorts of malignant mesothelioma patients, receiving tremelimumab as a single agent (NCT01843374), liver cancer patients, who are treated with a combination of tremelimumab and trans-arterial catheter chemoembolization or radiofrequency ablation (NCT01853618), and subjects affected by various solid neoplasms, who are given tremelimumab plus MEDI4736 (a human IgG1 specific for PD-L1) (NCT01975831). Of note, MEDI4736 as well as two other PD-L1 targeting mAbs, namely MPDL3280A (a human IgG1 also known as RG7446) and MSB0010718C, are being investigated also as standalone therapeutic interventions in cohorts of patients affected by NSCLC (NCT01846416; NCT01903993) or various advanced solid neoplasms (NCT01772004; NCT01938612; NCT01943461) (Table 3).
Table 3. Clinical trials recently launched to evaluate the therapeutic profile of immunostimulatory mAbs.*.
mAb | Target(s) | Indication(s) | Phase | Status | Note | Ref. |
---|---|---|---|---|---|---|
Anti-OX40 | TNFRSF4 (OX40) |
Breast carcinoma | I/II | Recruiting | Combined with SBRT | NCT01862900 |
Ipilimumab | CTLA4 | AML MDS |
I | Recruiting | As single agent | NCT01757639 |
Cervical carcinoma |
I | Recruiting | Combined with cisplatin and radiation therapy |
NCT01711515 | ||
CRC Lymphoma Melanoma |
I/II | Recruiting | Combined with radiation therapy | NCT01769222 | ||
Hematological malignancies | I | Recruiting | As single agent | NCT01822509 | ||
HNC | I | Recruiting | Combined with cetuximab and radiation therapy |
NCT01860430 | ||
I | Recruiting | Combined with cetuximab and radiation therapy |
NCT01935921 | |||
Hodgkin's lymphoma |
I | Not yet recruiting |
Combined with brentuximab vedotin |
NCT01896999 | ||
Leukemia Lymphoma |
n.a. | Not yet recruiting |
Combined with lenalidomide | NCT01919619 | ||
Lymphoma | I | Recruiting | Combined with rituximab | NCT01729806 | ||
Melanoma | n.a. | Not yet recruiting |
As single agent | NCT01715077 | ||
n.a. | Not yet recruiting |
In the context of 99Tc-based imaging procedures |
NCT01789827 | |||
n.a. | Recruiting | Followed by lymphodepletion, TIL infusion and IL-2 |
NCT01701674 | |||
0 | Recruiting | Combined with radioembolization | NCT01730157 | |||
I | Recruiting | Combined with SRS or WBRT | NCT01703507 | |||
I | Recruiting | Combined with dabrafenib ± trametinib | NCT01767454 | |||
I | Recruiting | Combined with NY-ESO-1-targeting vaccine ± montanide | NCT01810016 | |||
I | Recruiting | Combined with BCG | NCT01838200 | |||
I/II | Recruiting | As single agent or combined with oncolytic virotherapy |
NCT01740297 | |||
II | Active not recruiting |
Combined with cyclophosphamide | NCT01740401 | |||
II | Not yet recruiting |
As single agent | NCT01879306 | |||
II | Recruiting | As single agent or combined with IFNα-2b |
NCT01708941 | |||
II | Recruiting | As single agent | NCT01709162 | |||
II | Recruiting | Combined with nivolumab | NCT01783938 | |||
II | Recruiting | Combined with paclitaxel | NCT01827111 | |||
III | Recruiting | Combined with nivolumab | NCT01844505 | |||
III | Recruiting | Combined with lambrolizumab | NCT01866319 | |||
IV | Recruiting | Combined with high-dose IL-2 | NCT01856023 | |||
Merkel cell carcinoma |
II | Not yet recruiting |
As single agent | NCT01913691 | ||
NSCLC | I | Recruiting | Combined with carboplatin, cisplatin and paclitaxel | NCT01820754 | ||
Pancreatic cancer | II | Not yet recruiting |
As single agent or combined with a GM-CSF-secreting vaccine |
NCT01896869 | ||
Prostate cancer | I | Recruiting | Combined with sipuleucel-T | NCT01832870 | ||
II | Active not recruiting |
After sipuleucel-T treatment | NCT01804465 | |||
Advanced solid tumors |
I | Recruiting | Combined with imatinib | NCT01738139 | ||
I | Recruiting | Combined with lirilumab | NCT01750580 | |||
I | Recruiting | Combined with lenalidomide | NCT01750983 | |||
I/II | Not yet recruiting |
Combined with nivolumab | NCT01928394 | |||
Lambrolizumab | PDCD1 (PD-1, CD279) |
CRC | II | Recruiting | As single agent | NCT01876511 |
Hematological malignancies | I | Not yet recruiting |
As single agent | NCT01953692 | ||
Melanoma | II | Active not recruiting |
As single agent | NCT01704287 | ||
III | Recruiting | Combined with ipilimumab | NCT01866319 | |||
NSCLC | II/III | Recruiting | As single agent | NCT01905657 | ||
Advanced solid tumors |
I | Recruiting | As single agent or combined with conventional chemotherapy | NCT01840579 | ||
I | Recruiting | As single agent | NCT01848834 | |||
Lirilumab | KIR | Advanced solid tumors | I | Recruiting | Combined with nivolumab | NCT01714739 |
I | Recruiting | Combined with ipilimumab | NCT01750580 | |||
MEDI4736 | CD274 (PD-L1, B7-H1) |
Advanced solid tumors | I | Not yet recruiting |
Combined with tremelimumab | NCT01975831 |
I | Recruiting | As single agent | NCT01938612 | |||
MPDL3280A | CD274 (PD-L1, B7-H1) |
NSCLC | II | Recruiting | As single agent | NCT01846416 |
II | Recruiting | As single agent | NCT01903993 | |||
MSB0010718C | CD274 (PD-L1, B7-H1) |
Advanced solid tumors | I | Recruiting | As single agent | NCT01772004 |
I | Recruiting | As single agent | NCT01943461 | |||
Nivolumab | PDCD1 (PD-1, CD279) |
Melanoma | II | Recruiting | Combined with ipilimumab | NCT01783938 |
II | Recruiting | Combined with ipilimumab | NCT01927419 | |||
III | Recruiting | As single agent | NCT01721746 | |||
III | Recruiting | As single agent | NCT01721772 | |||
III | Recruiting | Combined with ipilimumab | NCT01844505 | |||
NSCLC | II | Active not recruiting |
As single agent | NCT01721759 | ||
II | Recruiting | Combined with azacitidine ± entinostat | NCT01928576 | |||
Advanced solid tumors | I | Recruiting | Combined with lirilumab | NCT01714739 | ||
I | Recruiting | As single agent or combined with immunotherapy |
NCT01968109 | |||
I/II | Not yet recruiting |
As single agent or combined with ipilimumab |
NCT01928394 | |||
Pidilizumab | PDCD1 (PD-1, CD279) |
Glioma | I/II | Not yet recruiting |
As single agent | NCT01952769 |
Tremelimumab | CTLA4 | HCC | I | Recruiting | Combined with RFA and TACE | NCT01853618 |
Mesothelioma | II | Recruiting | As single agent | NCT01843374 | ||
Advanced solid tumors | I | Not yet recruiting |
Combined with MEDI4736 | NCT01975831 | ||
Urelumab | TNFRSF9 (CD137, 4–1BB) |
CLL NHL |
I | Recruiting | Combined with rituximab | NCT01775631 |
Abbreviations: AML, acute myeloid leukemia; BCG, bacillus Calmette-Guérin; CLL, chronic lymphocytic leukemia; CRC, colorectal carcinoma; CTLA4, cytotoxic T lymphocyte-associated protein 4; GM-CSF, granulocyte macrophage colony-stimulating factor; HCC, hepatocellular carcinoma; HNC, head and neck cancer; IFNα-2b, interferon α-2b; IL, interleukin; KIR, killer cell immunoglobulin-like receptor; mAb, monoclonal antibody; MDS, myelodysplastic syndrome; n.a., not available; NHL, Non-Hodgkin's lymphoma; NSCLC, non-small cell lung carcinoma; PDCD1, programmed cell death 1; RFA, radiofrequency ablation; SBRT, stereotactic body radiation therapy; SRS, stereotactic radiosurgery; TACE, transarterial catheter chemoembolization; TIL, tumor-infiltrating lymphocyte; TNFRSF, tumor necrosis factor receptor superfamily; WBRT; whole-brain radiation therapy. *between 2012, October 1st and the day of submission.
As a matter of fact, targeting PD-1-dependent immunosuppression currently stands out as the experimental mAb-based immunotherapeutic paradigm most intensively investigated in clinical settings. Thus, nivolumab is being tested, most often as a standalone intervention or combined with ipilimumab, in patients with melanoma (NCT01721746; NCT01721772; NCT01783938; NCT01844505; NCT01927419), NLSLC (NCT01721759; NCT01928576), glioma (NCT01952769) or other solid neoplasms (NCT01714739; NCT01928394; NCT01968109). Alongside, the safety and therapeutic profile of lambrolizumab, most frequently employed as a single agent or together with ipilimumab, are being assessed in cohorts of subjects bearing melanoma (NCT01704287; NCT01866319), NSCLC (NCT01840579; NCT01905657), hematological malignancies (NCT01953692), colorectal carcinoma with high degrees of microsatellite instability (NCT01876511), or advanced/metastatic tumors (NCT01840579; NCT01848834) (Table 3).
Finally, some interest is growing around the possibility to harness the antineoplastic activity of NK cell-targeting mAbs as well as mAbs that operate as agonists for co-stimulatory T-cell and NK-cell receptors. In this setting, lirilumab (a human IgG4 that antagonizes KIRs, also known as IPH2102)109 is being tested in combination with ipilimumab or nivolumab for the treatment of patients bearing advanced solid malignancies (NCT01714739; NCT01750580). Moreover, the safety and therapeutic potential of the TNFRSF9-activating mAb urelumab110 and a not better specified OX40-activating mAb, combined with rituximab and stereotactic body radiation, respectively,6,7,111 are being assessed in cohorts of patients with CLL, NHL (NCT01775631) or metastatic breast carcinoma (NCT01862900) (Table 3).
As for the clinical trials listed in our previous Trial Watches dealing with this topic,6,7 only NCT01034787 has changed status. As a matter of fact, the status of NCT01034787, a Phase 2 trial testing intravenous tremelimumab in patients affected by uveal melanoma, is no longer available because the information relative to this study has not been verified recently (source http://www.clinicaltrials.gov).
Concluding Remarks
Similar to their tumor-targeting counterparts,8 immunostimulatory mAbs are being intensively investigated for their ability to mediate therapeutically relevant antineoplastic effects. However, while tumor-targeting mAbs are designed to specifically bind to malignant, stromal or endothelial components of neoplastic lesions or to neutralize trophic signals delivered by the tumor stroma,1,2 most immunostimulatory mAbs modulate general mechanisms that control innate and adaptive immune responses, de facto operating in a relatively unspecific manner.3,4
Based on this consideration, one would expect immunostimulatory mAbs to provoke more severe side effects than their tumor-targeting counterparts3,4 and to display a therapeutic activity resembling that of other relatively unspecific immunostimulatory interventions, such as TLR agonists,104,105,112,113 specific cytokines,102,103 and immunogenic chemotherapeutics.74,75,114-116 Accumulating clinical evidence actually suggests that immunostimulatory mAbs are generally well tolerated by cancer patients, in particular when administered as standalone therapeutic interventions.3,4 Conversely, the true clinical potential of most immunostimulatory mAbs employed as single agents remains matter of debate. So far, ipilimumab (the sole immunostimulatory mAb currently approved for use in humans) has been shown to exert robust therapeutic effects only in patients affected by melanoma,58-60 a type of tumor that (1) is considered as inherently immunogenic, and (2) at least in part, responds to other unspecific immunostimulants, including high-dose interleukin-2 and interferon α2b.102,103 Moreover, the clinical efficacy of ipilimumab seems a unique prerogative of this mAb rather than a general feature of CTL4-targeting mAbs such as tremelimumab.84 The development of immunostimulatory mAbs is in its infancy and further studies are required to obtain profound insights into the pharmacodynamics and pharmacokinetic properties of molecules that are conceived to target the same immunomodulatory receptor yet exhibit differential clinical activity. As it stands, immunomodulatory mAbs hold great promise as tools to potentiate tumor-targeting immune responses elicited by active immunotherapeutic interventions, including recombinant vaccines,117,118 dendritic cell-based approaches,100,101,119,120 and adoptive T-cell transfer.121-124 The future will tell whether immunostimulatory mAbs other than ipilimumab will add to the growing list of FDA-approved anticancer immunotherapeutics.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Acknowledgments
Authors are supported by the Ligue contre le Cancer (équipe labelisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt–Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).
Glossary
Abbreviations:
- CLL
chronic lymphocytic leukemia
- CTLA4
cytotoxic T lymphocyte-associated protein 4
- FDA
Food and Drug Administration
- GM-CSF
granulocyte macrophage colony-stimulating factor
- KIR
killer cell immunoglobulin-like receptor
- mAb
monoclonal antibody
- NHL
non-Hodgkin’s lymphoma
- NK
natural killer
- NSCLC
non-small cell lung carcinoma
- TNFRSF
tumor necrosis factor receptor superfamily, member
- TLR
Toll-like receptor
Citation: Aranda F, Eggermont A, Vacchelli E, Galon J, Chernyavsky A, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch - Immunostimulatory monoclonal antibodies in cancer therapy. OncoImmunology 2014; 3:e27297; 10.4161/onci.27297
Footnotes
Previously published online: www.landesbioscience.com/journals/oncoimmunology/article/27297
References
- 1.Alkan SS. Monoclonal antibodies: the story of a discovery that revolutionized science and medicine. Nat Rev Immunol. 2004;4:153–6. doi: 10.1038/nri1265. [DOI] [PubMed] [Google Scholar]
- 2.Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol. 2010;10:317–27. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Melero I, Grimaldi AM, Perez-Gracia JL, Ascierto PA. Clinical development of immunostimulatory monoclonal antibodies and opportunities for combination. Clin Cancer Res. 2013;19:997–1008. doi: 10.1158/1078-0432.CCR-12-2214. [DOI] [PubMed] [Google Scholar]
- 4.Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer. 2007;7:95–106. doi: 10.1038/nrc2051. [DOI] [PubMed] [Google Scholar]
- 5.Gray JC, Johnson PW, Glennie MJ. Therapeutic potential of immunostimulatory monoclonal antibodies. Clin Sci (Lond) 2006;111:93–106. doi: 10.1042/CS20060024. [DOI] [PubMed] [Google Scholar]
- 6.Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi: 10.4161/onci.1.1.17938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2013;2:e22789. doi: 10.4161/onci.22789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Vacchelli E, Aranda F, Eggermont A, Galon J, Sautes-Fridman C, Zitvogel L, et al. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. OncoImmunology. 2014 doi: 10.4161/onci.27048. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391–400. doi: 10.1038/nrd1381. [DOI] [PubMed] [Google Scholar]
- 10.Michielsen AJ, Ryan EJ, O’Sullivan JN. Dendritic cell inhibition correlates with survival of colorectal cancer patients on bevacizumab treatment. Oncoimmunology. 2012;1:1445–7. doi: 10.4161/onci.21318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, Morel P, Van Den Neste E, Salles G, Gaulard P, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med. 2002;346:235–42. doi: 10.1056/NEJMoa011795. [DOI] [PubMed] [Google Scholar]
- 12.McLaughlin P, Grillo-López AJ, Link BK, Levy R, Czuczman MS, Williams ME, Heyman MR, Bence-Bruckler I, White CA, Cabanillas F, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol. 1998;16:2825–33. doi: 10.1200/JCO.1998.16.8.2825. [DOI] [PubMed] [Google Scholar]
- 13.Sorbye SW, Kilvaer T, Valkov A, Donnem T, Smeland E, Al-Shibli K, Bremnes RM, Busund LT. High expression of CD20+ lymphocytes in soft tissue sarcomas is a positive prognostic indicator. Oncoimmunology. 2012;1:75–7. doi: 10.4161/onci.1.1.17825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24:19–28. doi: 10.1016/j.immuni.2005.11.010. [DOI] [PubMed] [Google Scholar]
- 15.Hubert P, Amigorena S. Antibody-dependent cell cytotoxicity in monoclonal antibody-mediated tumor immunotherapy. Oncoimmunology. 2012;1:103–5. doi: 10.4161/onci.1.1.17963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Houot R, Kohrt H, Levy R. Boosting antibody-dependant cellular cytotoxicity against tumor cells with a CD137 stimulatory antibody. Oncoimmunology. 2012;1:957–8. doi: 10.4161/onci.19974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kute T, Stehle JR, Jr., Ornelles D, Walker N, Delbono O, Vaughn JP. Understanding key assay parameters that affect measurements of trastuzumab-mediated ADCC against Her2 positive breast cancer cells. Oncoimmunology. 2012;1:810–21. doi: 10.4161/onci.20447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Winiarska M, Glodkowska-Mrowka E, Bil J, Golab J. Molecular mechanisms of the antitumor effects of anti-CD20 antibodies. Front Biosci (Landmark Ed) 2011;16:277–306. doi: 10.2741/3688. [DOI] [PubMed] [Google Scholar]
- 19.Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res. 2010;20:34–50. doi: 10.1038/cr.2009.139. [DOI] [PubMed] [Google Scholar]
- 20.Zipfel PF, Skerka C. Complement regulators and inhibitory proteins. Nat Rev Immunol. 2009;9:729–40. doi: 10.1038/nri2620. [DOI] [PubMed] [Google Scholar]
- 21.Weiner LM, Belldegrun AS, Crawford J, Tolcher AW, Lockbaum P, Arends RH, Navale L, Amado RG, Schwab G, Figlin RA. Dose and schedule study of panitumumab monotherapy in patients with advanced solid malignancies. Clin Cancer Res. 2008;14:502–8. doi: 10.1158/1078-0432.CCR-07-1509. [DOI] [PubMed] [Google Scholar]
- 22.Ming Lim C, Stephenson R, Salazar AM, Ferris RL. TLR3 agonists improve the immunostimulatory potential of cetuximab against EGFR(+) head and neck cancer cells. Oncoimmunology. 2013;2:e24677. doi: 10.4161/onci.24677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kawaguchi Y, Kono K, Mimura K, Sugai H, Akaike H, Fujii H. Cetuximab induce antibody-dependent cellular cytotoxicity against EGFR-expressing esophageal squamous cell carcinoma. Int J Cancer. 2007;120:781–7. doi: 10.1002/ijc.22370. [DOI] [PubMed] [Google Scholar]
- 24.Srivastava RM, Lee SC, Andrade Filho PA, Lord CA, Jie HB, Davidson HC, López-Albaitero A, Gibson SP, Gooding WE, Ferrone S, et al. Cetuximab-activated natural killer and dendritic cells collaborate to trigger tumor antigen-specific T-cell immunity in head and neck cancer patients. Clin Cancer Res. 2013;19:1858–72. doi: 10.1158/1078-0432.CCR-12-2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
- 26.Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39:74–88. doi: 10.1016/j.immuni.2013.06.014. [DOI] [PubMed] [Google Scholar]
- 27.Witzig TE, Gordon LI, Cabanillas F, Czuczman MS, Emmanouilides C, Joyce R, Pohlman BL, Bartlett NL, Wiseman GA, Padre N, et al. Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma. J Clin Oncol. 2002;20:2453–63. doi: 10.1200/JCO.2002.11.076. [DOI] [PubMed] [Google Scholar]
- 28.Kaminski MS, Estes J, Zasadny KR, Francis IR, Ross CW, Tuck M, Regan D, Fisher S, Gutierrez J, Kroll S, et al. Radioimmunotherapy with iodine (131)I tositumomab for relapsed or refractory B-cell non-Hodgkin lymphoma: updated results and long-term follow-up of the University of Michigan experience. Blood. 2000;96:1259–66. [PubMed] [Google Scholar]
- 29.Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol. 2011;11:852–63. doi: 10.1038/nri3108. [DOI] [PubMed] [Google Scholar]
- 30.Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4:336–47. doi: 10.1038/nri1349. [DOI] [PubMed] [Google Scholar]
- 31.Waitz R, Fassò M, Allison JP. CTLA-4 blockade synergizes with cryoablation to mediate tumor rejection. Oncoimmunology. 2012;1:544–6. doi: 10.4161/onci.19442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Munir S, Andersen GH, Svane IM, Andersen MH. The immune checkpoint regulator PD-L1 is a specific target for naturally occurring CD4(+) T cells. Oncoimmunology. 2013;2:e23991. doi: 10.4161/onci.23991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology. 2012;1:1223–5. doi: 10.4161/onci.21335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, Azuma M, Krummel MF, Bluestone JA. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol. 2009;10:1185–92. doi: 10.1038/ni.1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.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;8:239–45. doi: 10.1038/ni1443. [DOI] [PubMed] [Google Scholar]
- 36.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–8. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 37.Raulet DH, Guerra N. Oncogenic stress sensed by the immune system: role of natural killer cell receptors. Nat Rev Immunol. 2009;9:568–80. doi: 10.1038/nri2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Smyth MJ, Hayakawa Y, Takeda K, Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat Rev Cancer. 2002;2:850–61. doi: 10.1038/nrc928. [DOI] [PubMed] [Google Scholar]
- 39.Joncker NT, Raulet DH. Regulation of NK cell responsiveness to achieve self-tolerance and maximal responses to diseased target cells. Immunol Rev. 2008;224:85–97. doi: 10.1111/j.1600-065X.2008.00658.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Long EO. Negative signaling by inhibitory receptors: the NK cell paradigm. Immunol Rev. 2008;224:70–84. doi: 10.1111/j.1600-065X.2008.00660.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Withers DR, Gaspal FM, Bekiaris V, McConnell FM, Kim M, Anderson G, Lane PJ. OX40 and CD30 signals in CD4(+) T-cell effector and memory function: a distinct role for lymphoid tissue inducer cells in maintaining CD4(+) T-cell memory but not effector function. Immunol Rev. 2011;244:134–48. doi: 10.1111/j.1600-065X.2011.01057.x. [DOI] [PubMed] [Google Scholar]
- 42.Croft M. The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol. 2009;9:271–85. doi: 10.1038/nri2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sugamura K, Ishii N, Weinberg AD. Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40. Nat Rev Immunol. 2004;4:420–31. doi: 10.1038/nri1371. [DOI] [PubMed] [Google Scholar]
- 44.Hombach AA, Heiders J, Foppe M, Chmielewski M, Abken H. OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4(+) T cells. Oncoimmunology. 2012;1:458–66. doi: 10.4161/onci.19855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Shevach EM, Stephens GL. The GITR-GITRL interaction: co-stimulation or contrasuppression of regulatory activity? Nat Rev Immunol. 2006;6:613–8. doi: 10.1038/nri1867. [DOI] [PubMed] [Google Scholar]
- 46.Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S. Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol. 2002;3:135–42. doi: 10.1038/ni759. [DOI] [PubMed] [Google Scholar]
- 47.Chen W, Frank ME, Jin W, Wahl SM. TGF-beta released by apoptotic T cells contributes to an immunosuppressive milieu. Immunity. 2001;14:715–25. doi: 10.1016/S1074-7613(01)00147-9. [DOI] [PubMed] [Google Scholar]
- 48.Schnurr M, Duewell P. Breaking tumor-induced immunosuppression with 5′-triphosphate siRNA silencing TGFβ and activating RIG-I. Oncoimmunology. 2013;2:e24170. doi: 10.4161/onci.24170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell biology. Nat Rev Immunol. 2002;2:46–53. doi: 10.1038/nri704. [DOI] [PubMed] [Google Scholar]
- 50.Pickup M, Novitskiy S, Moses HL. The roles of TGFβ in the tumour microenvironment. Nat Rev Cancer. 2013;13:788–99. doi: 10.1038/nrc3603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–24. doi: 10.1038/nrc2853. [DOI] [PubMed] [Google Scholar]
- 52.LoBuglio AF, Saleh MN, Lee J, Khazaeli MB, Carrano R, Holden H, Wheeler RH. Phase I trial of multiple large doses of murine monoclonal antibody CO17-1A. I. Clinical aspects. J Natl Cancer Inst. 1988;80:932–6. doi: 10.1093/jnci/80.12.932. [DOI] [PubMed] [Google Scholar]
- 53.Lobuglio AF, Saleh M, Peterson L, Wheeler R, Carrano R, Huster W, Khazaeli MB. Phase I clinical trial of CO17-1A monoclonal antibody. Hybridoma. 1986;5(Suppl 1):S117–23. [PubMed] [Google Scholar]
- 54.Sears HF, Herlyn D, Steplewski Z, Koprowski H. Phase II clinical trial of a murine monoclonal antibody cytotoxic for gastrointestinal adenocarcinoma. Cancer Res. 1985;45:5910–3. [PubMed] [Google Scholar]
- 55.Sears HF, Atkinson B, Mattis J, Ernst C, Herlyn D, Steplewski Z, Häyry P, Koprowski H. Phase-I clinical trial of monoclonal antibody in treatment of gastrointestinal tumours. Lancet. 1982;1:762–5. doi: 10.1016/S0140-6736(82)91811-6. [DOI] [PubMed] [Google Scholar]
- 56.Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
- 57.Matzinger P. The danger model: a renewed sense of self. Science. 2002;296:301–5. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
- 58.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Robert C, Thomas L, Bondarenko I, O’Day S, M D JW, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
- 60.Wolchok JD, Neyns B, Linette G, Negrier S, Lutzky J, Thomas L, Waterfield W, Schadendorf D, Smylie M, Guthrie T, Jr., et al. Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol. 2010;11:155–64. doi: 10.1016/S1470-2045(09)70334-1. [DOI] [PubMed] [Google Scholar]
- 61.Sosman J, Sznol M, McDermott D, Carvajal RD, Lawrence DP, Topalian SL, et al. Clinical activity and safety of anti-programmed death-1 (PD-1) (BMS-936558/MDX-1106/ONO-4538) in patients (pts) with advanced melanoma (mel) Ann Oncol. 2012;23: ix367–75. [Google Scholar]
- 62.Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75. doi: 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Patnaik A, Kang SP, Tolcher AW, Rasco DW, Papadopoulos KP, Beeram M, et al. Phase I study of MK-3475 (anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. . J Clin Oncol. 2012;30:abstr 2512. doi: 10.1158/1078-0432.CCR-14-2607. [DOI] [PubMed] [Google Scholar]
- 66.Tykodi SS, Brahmer JR, Hwu WJ, Chow LQ, Hwu P, Odunsi K, et al. PD-1/PD-L1 pathway as a target for cancer immunotherapy: safety and clinical activity of BMS-936559, an anti-PD-L1 antibody, in patients with solid tumors. J Clin Oncol. 2012;30:abstr 2510. [Google Scholar]
- 67.Vonderheide RH, Flaherty KT, Khalil M, Stumacher MS, Bajor DL, Hutnick NA, Sullivan P, Mahany JJ, Gallagher M, Kramer A, 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;25:876–83. doi: 10.1200/JCO.2006.08.3311. [DOI] [PubMed] [Google Scholar]
- 68.Hussein M, Berenson JR, Niesvizky R, Munshi N, Matous J, Sobecks R, Harrop K, Drachman JG, Whiting N. A phase I multidose study of dacetuzumab (SGN-40; humanized anti-CD40 monoclonal antibody) in patients with multiple myeloma. Haematologica. 2010;95:845–8. doi: 10.3324/haematol.2009.008003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.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;51:228–35. doi: 10.3109/10428190903440946. [DOI] [PubMed] [Google Scholar]
- 70.Ascierto PA, Simeone E, Sznol M, Fu YX, Melero I. Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol. 2010;37:508–16. doi: 10.1053/j.seminoncol.2010.09.008. [DOI] [PubMed] [Google Scholar]
- 71.Molckovsky A, Siu 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]
- 72.Aranda F, Vacchelli E, Eggermont A, Galon J, Sautes-Fridman C, Tartour E, et al. Trial Watch: Peptide vaccines in cancer therapy. OncoImmunology. 2013;2:e26621. doi: 10.4161/onci.26621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology. 2012;1:1557–76. doi: 10.4161/onci.22428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Kroemer G. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Vacchelli E, Senovilla L, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2013;2:e23510. doi: 10.4161/onci.23510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Sherrill B, Wang J, Kotapati S, Chin K. Q-TWiST analysis comparing ipilimumab/dacarbazine vs placebo/dacarbazine for patients with stage III/IV melanoma. Br J Cancer. 2013;109:8–13. doi: 10.1038/bjc.2013.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Santegoets SJ, Stam AG, Lougheed SM, Gall H, Scholten PE, Reijm M, Jooss K, Sacks N, Hege K, Lowy I, et al. T cell profiling reveals high CD4+CTLA-4 + T cell frequency as dominant predictor for survival after prostate GVAX/ipilimumab treatment. Cancer Immunol Immunother. 2013;62:245–56. doi: 10.1007/s00262-012-1330-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ribas A, Hodi FS, Callahan M, Konto C, Wolchok J. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med. 2013;368:1365–6. doi: 10.1056/NEJMc1302338. [DOI] [PubMed] [Google Scholar]
- 79.Reck M, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, Sebastian M, Lu H, Cuillerot JM, Lynch TJ. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol. 2013;24:75–83. doi: 10.1093/annonc/mds213. [DOI] [PubMed] [Google Scholar]
- 80.Le DT, Lutz E, Uram JN, Sugar EA, Onners B, Solt S, Zheng L, Diaz LA, Jr., Donehower RC, Jaffee EM, et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J Immunother. 2013;36:382–9. doi: 10.1097/CJI.0b013e31829fb7a2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kwek SS, Dao V, Roy R, Hou Y, Alajajian D, Simko JP, Small EJ, Fong L. Diversity of antigen-specific responses induced in vivo with CTLA-4 blockade in prostate cancer patients. J Immunol. 2012;189:3759–66. doi: 10.4049/jimmunol.1201529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–33. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ribas A, Chesney JA, Gordon MS, Abernethy AP, Logan TF, Lawson DH, Chmielowksi B, Glaspy JA, Lewis K, Huang B, et al. Safety profile and pharmacokinetic analyses of the anti-CTLA4 antibody tremelimumab administered as a one hour infusion. J Transl Med. 2012;10:236. doi: 10.1186/1479-5876-10-236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ribas A, Kefford R, Marshall MA, Punt CJ, Haanen JB, Marmol M, Garbe C, Gogas H, Schachter J, Linette G, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol. 2013;31:616–22. doi: 10.1200/JCO.2012.44.6112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Millward M, Underhill C, Lobb S, McBurnie J, Meech SJ, Gomez-Navarro J, Marshall MA, Huang B, Mather CB. Phase I study of tremelimumab (CP-675 206) plus PF-3512676 (CPG 7909) in patients with melanoma or advanced solid tumours. Br J Cancer. 2013;108:1998–2004. doi: 10.1038/bjc.2013.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lipson EJ, Sharfman WH, Drake CG, Wollner I, Taube JM, Anders RA, Xu H, Yao S, Pons A, Chen L, et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res. 2013;19:462–8. doi: 10.1158/1078-0432.CCR-12-2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–44. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Vey N, Bourhis JH, Boissel N, Bordessoule D, Prebet T, Charbonnier A, Etienne A, Andre P, Romagne F, Benson D, et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood. 2012;120:4317–23. doi: 10.1182/blood-2012-06-437558. [DOI] [PubMed] [Google Scholar]
- 89.Benson DM, Jr., Hofmeister CC, Padmanabhan S, Suvannasankha A, Jagannath S, Abonour R, Bakan C, Andre P, Efebera Y, Tiollier J, et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood. 2012;120:4324–33. doi: 10.1182/blood-2012-06-438028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Vonderheide RH, Burg JM, Mick R, Trosko JA, Li D, Shaik MN, Tolcher AW, Hamid O. Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors. Oncoimmunology. 2013;2:e23033. doi: 10.4161/onci.23033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Forero-Torres A, Bartlett N, Beaven A, Myint H, Nasta S, Northfelt DW, Whiting NC, Drachman JG, Lobuglio AF, Moskowitz CH. Pilot study of dacetuzumab in combination with rituximab and gemcitabine for relapsed or refractory diffuse large B-cell lymphoma. Leuk Lymphoma. 2013;54:277–83. doi: 10.3109/10428194.2012.710328. [DOI] [PubMed] [Google Scholar]
- 92.Beatty GL, Torigian DA, Chiorean EG, Saboury B, Brothers A, Alavi A, Troxel AB, Sun W, Teitelbaum UR, Vonderheide RH, et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res. 2013;19:6286–95. doi: 10.1158/1078-0432.CCR-13-1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Byrd JC, Kipps TJ, Flinn IW, Cooper M, Odenike O, Bendiske J, Rediske J, Bilic S, Dey J, Baeck J, et al. Phase I study of the anti-CD40 humanized monoclonal antibody lucatumumab (HCD122) in relapsed chronic lymphocytic leukemia. Leuk Lymphoma. 2012;53:2136–42. doi: 10.3109/10428194.2012.681655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K, Walker J, Gonzalez I, Meeuwsen T, Fox BA, et al. OX40 Is a Potent Immune-Stimulating Target in Late-Stage Cancer Patients. Cancer Res. 2013;73:7189–98. doi: 10.1158/0008-5472.CAN-12-4174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wang-Gillam A, Plambeck-Suess S, Goedegebuure P, Simon PO, Mitchem JB, Hornick JR, Sorscher S, Picus J, Suresh R, Lockhart AC, et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Invest New Drugs. 2013;31:707–13. doi: 10.1007/s10637-012-9866-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hoffmann J, Vitale I, Buchmann B, Galluzzi L, Schwede W, Senovilla L, Skuballa W, Vivet S, Lichtner RB, Vicencio JM, et al. Improved cellular pharmacokinetics and pharmacodynamics underlie the wide anticancer activity of sagopilone. Cancer Res. 2008;68:5301–8. doi: 10.1158/0008-5472.CAN-08-0237. [DOI] [PubMed] [Google Scholar]
- 97.Senovilla L, Vitale I, Martins I, Tailler M, Pailleret C, Michaud M, Galluzzi L, Adjemian S, Kepp O, Niso-Santano M, et al. An immunosurveillance mechanism controls cancer cell ploidy. Science. 2012;337:1678–84. doi: 10.1126/science.1224922. [DOI] [PubMed] [Google Scholar]
- 98.Michels J, Vitale I, Galluzzi L, Adam J, Olaussen KA, Kepp O, Senovilla L, Talhaoui I, Guegan J, Enot DP, et al. Cisplatin resistance associated with PARP hyperactivation. Cancer Res. 2013;73:2271–80. doi: 10.1158/0008-5472.CAN-12-3000. [DOI] [PubMed] [Google Scholar]
- 99.Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G. Molecular mechanisms of cisplatin resistance. Oncogene. 2012;31:1869–83. doi: 10.1038/onc.2011.384. [DOI] [PubMed] [Google Scholar]
- 100.Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1:1111–34. doi: 10.4161/onci.21494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2013;2:e25771. doi: 10.4161/onci.25771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Vacchelli E, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory cytokines. Oncoimmunology. 2013;2:e24850. doi: 10.4161/onci.24850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Immunostimulatory cytokines. Oncoimmunology. 2012;1:493–506. doi: 10.4161/onci.20459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:699–716. doi: 10.4161/onci.20696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:894–907. doi: 10.4161/onci.20931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Vacchelli E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2013;2:e25238. doi: 10.4161/onci.25238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Tarhini AA. Tremelimumab: a review of development to date in solid tumors. Immunotherapy. 2013;5:215–29. doi: 10.2217/imt.13.9. [DOI] [PubMed] [Google Scholar]
- 108.Bensinger W, Maziarz RT, Jagannath S, Spencer A, Durrant S, Becker PS, Ewald B, Bilic S, Rediske J, Baeck J, 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;159:58–66. doi: 10.1111/j.1365-2141.2012.09251.x. [DOI] [PubMed] [Google Scholar]
- 109.Alici E. IPH-2101, a fully human anti-NK-cell inhibitory receptor mAb for the potential treatment of hematological cancers. Curr Opin Mol Ther. 2010;12:724–33. [PubMed] [Google Scholar]
- 110.Chacon JA, Wu RC, Sukhumalchandra P, Molldrem JJ, Sarnaik A, Pilon-Thomas S, Weber J, Hwu P, Radvanyi L. Co-stimulation through 4-1BB/CD137 improves the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy. PLoS One. 2013;8:e60031. doi: 10.1371/journal.pone.0060031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Anticancer radioimmunotherapy. Oncoimmunology. 2013;2:e25595. doi: 10.4161/onci.25595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013;59:583–94. doi: 10.1016/j.jhep.2013.03.033. [DOI] [PubMed] [Google Scholar]
- 113.Rakoff-Nahoum S, Medzhitov R. Toll-like receptors and cancer. Nat Rev Cancer. 2009;9:57–63. doi: 10.1038/nrc2541. [DOI] [PubMed] [Google Scholar]
- 114.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
- 115.Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12:860–75. doi: 10.1038/nrc3380. [DOI] [PubMed] [Google Scholar]
- 116.Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, Kroemer G, et al. Trial Watch: Lenalidomide-based immunochemotherapy. OncoImmunology. 2013;2:e26494. doi: 10.4161/onci.26494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Stevanovic S. Identification of tumour-associated T-cell epitopes for vaccine development. Nat Rev Cancer. 2002;2:514–20. doi: 10.1038/nrc841. [DOI] [PubMed] [Google Scholar]
- 118.Eggermont AM. Immunotherapy: Vaccine trials in melanoma -- time for reflection. Nat Rev Clin Oncol. 2009;6:256–8. doi: 10.1038/nrclinonc.2009.42. [DOI] [PubMed] [Google Scholar]
- 119.Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449:419–26. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
- 120.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12:265–77. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Vacchelli E, Eggermont A, Fridman WH, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. Oncoimmunology. 2013;2:e24238. doi: 10.4161/onci.24238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology. 2012;1:306–15. doi: 10.4161/onci.19549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hinrichs CS, Restifo NP. Reassessing target antigens for adoptive T-cell therapy. Nat Biotechnol. 2013;31:999–1008. doi: 10.1038/nbt.2725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012;12:269–81. doi: 10.1038/nri3191. [DOI] [PMC free article] [PubMed] [Google Scholar]