1,659
Views
30
CrossRef citations to date
0
Altmetric
Review

Trial Watch

Adoptive cell transfer for anticancer immunotherapy

, , , , , , , , , & show all
Article: e28344 | Received 24 Feb 2014, Accepted 24 Feb 2014, Published online: 01 May 2014

Abstract

The expression “adoptive cell transfer” (ACT) is commonly employed to indicate an immunotherapeutic regimen involving the isolation of autologous blood-borne or tumor-infiltrating lymphocytes, their selection/expansion/activation ex vivo, and their reinfusion into the patient, most often in the context of lymphodepleting pre-conditioning and in combination with immunostimulatory treatments. Optionally, the cellular material for ACT is genetically manipulated before expansion to (1) target specific tumor-associated antigens; (2) endogenously express immunostimulatory molecules; and/or (3) persist for long periods upon reinfusion. Consistent efforts have been dedicated at the amelioration of this immunotherapeutic regimen throughout the past decade, resulting in the establishment of ever more efficient and safer ACT protocols. Accordingly, the number of clinical trials testing ACT in oncological indications does not cease to increase. In this Trial Watch, we summarize recent developments in this exciting area of research, covering both high-impact studies that have been published during the last 12 months and clinical trials that have been launched in the same period to evaluate the safety and therapeutic potential of ACT in cancer patients.

Introduction

The term “adoptive cell transfer” (ACT) is generally employed to identify a peculiar instance of anticancer immunotherapy relying on (1) the isolation of circulating or tumor-infiltrating lymphocytes; (2) their selection/expansion/activation ex vivo; and (3) their (re-)introduction into the patients, near-to-invariably in the context of lymphodepleting pre-conditioning and in combination with immunostimulatory agents.Citation1-Citation10 Thus, ACT should be conceptually differentiated from 2 other anticancer treatments relying on the (re)infusion of living cells, i.e., dendritic cell (DC)-based therapy and allogeneic hematopoietic stem cell transplantation (HSCT). Indeed, while ACT consists in the (re)infusion of autologous tumor-reactive lymphocytes, DC-based interventions (1) aim at (re)activating an endogenous tumor-specific immune response, de facto constituting anticancer vaccines; and (2) are never implemented upon lymphodepletion.Citation11-Citation16 Along similar lines, ACT cannot be compared with HSCT, a therapeutic option for patients affected by hematological neoplasms that involves (1) the eradication of a majority of malignant cells by lympho/myeloablating chemo(radio)therapy, and (2) the subsequent re-establishment of a healthy, allogeneic (and hence potentially tumor-reactive) immune system.Citation17,Citation18 Of note, although the therapeutic activity of ACT is generally ascribed to CD8+ T lymphocytes, the re-infusion of purified CD4+ T cells has been shown to yield durable clinical responses in melanoma patients.Citation19 Conversely, in spite of encouraging preclinical observations,Citation20-Citation26 the adoptive transfer of natural killer (NK) cells has been associated with a relatively deceiving clinical activity.Citation27-Citation29 Although strategies for endowing NK cells with MHC-independent, tumor-associated antigen (TAA)-specific reactivity may circumvent, at least in part, such a limitation,Citation30-Citation33 these approaches have not yet been tested in the clinic. Along similar lines, the actual therapeutic potential of B lymphocytes in cancer patients has not yet been investigated,Citation34 possibly because these cells reportedly exert immunosuppressive effects (at least in some circumstances).Citation35-Citation37

In oncological settings including melanoma and renal cell carcinoma (RCC), ACT relies on the reinfusion of tumor-infiltrating lymphocytes (TILs) that have been expanded ex vivo in the presence of growth factors such as interleukin (IL)-2 and (optionally) specific or unspecific activation stimuli.Citation38,Citation39 In most clinical scenarios, however, this strategy is unfeasible as (1) surgical/bioptic specimens for the isolation of TILs are not available, or (2) neoplastic lesions exhibit limited lymphocytic infiltration. Under these circumstances, ACT relies on peripheral blood lymphocytes (PBLs) that are endowed with tumor reactivity by genetic engineering.Citation40 Nowadays, this can be accomplished by 2 approaches. PBLs can indeed be manipulated to express a TAA-specific T-cell receptor (TCR),Citation40-Citation44 or a so-called chimeric antigen receptor (CAR), i.e., a transmembrane receptor consisting in the TAA-binding domain of an immunoglobulin fused to an intracellular tail containing 1 or more immunostimulatory signaling modules.Citation45-Citation51 As compared with TCRs, CARs are advantageous in that they allow adoptively transferred PBLs to recognize TAAs and exert robust cytotoxic functions in an MHC-independent fashion. In line with this notion, consistent rates of objective responses have been documented in cancer patients receiving CAR-engineered PBLs, especially in the case of hematological neoplasms.Citation49,Citation52-Citation58 Nonetheless, no protocol for ACT-based immunotherapy is currently approved by the US Food and Drug Administration (FDA) or other international regulatory agencies for use in cancer patients (source http://www.fda.gov).

Genetic engineering can also be employed to improve various aspects of the PBL biology prior to reinfusion, including proliferative capacity and in vivo persistence,Citation59-Citation62 secretory profile,Citation60 tumor-infiltrating potential,Citation63,Citation64 and cytotoxic activity.Citation65 Moreover, the cellular material for ACT can be derived in conditions that limit terminal differentiation, which is generally associated with at least some degree of proliferative impairment and/or functional exhaustion.Citation66-Citation68 Such a “rejuvenation” can be achieved with pharmacological modulators that promote the expansion of stem-cell memory T (TSCM) cells,Citation67,Citation69-Citation71 or via the induced pluripotent stem cell technology.Citation72-Citation74 Both these approaches allow indeed for the generation of TAA-specific T lymphocytes exhibiting improved persistence in vivo and cytotoxic functions as compared with tumor-reactive T cells obtained by conventional methods.Citation70-Citation74

ACT is generally performed in the contest of lymphodepleting regimens and combined with immunostimulatory interventions.Citation75-Citation77 Lymphodepletion has been consistently associated with an improvement in the clinical efficacy of ACT, presumably as (1) it relieves the potent immunosuppressive networks generally established in cancer patients by myeloid-derived suppressor cells (MDSCs)Citation78-Citation80 and FOXP3+ regulatory T cells;Citation81-Citation85 and (2) it reduces the size of the so-called “cytokine sink,” the compartment of endogenous immune effectors that may compete with re-infused lymphocytes for crucial cytokines such as IL-7 or IL-15.Citation86,Citation87 Along similar lines, various immunostimulatory agents have been shown to ameliorate the therapeutic profile of ACT, including (1) several cytokines, boosting the proliferative potential and/or cytotoxic functions of re-infused lymphocytes;Citation88-Citation91 (2) angiogenesis inhibitors, facilitating the homing of PBLs or TILs to malignant lesions;Citation92,Citation93 (3) Toll-like receptor ligands, operating as adjuvants;Citation94-Citation97 and (4) multiple chemotherapeutic agents that have been attributed a direct or indirect immunomodulatory activity.Citation98,Citation99

The major side effect of ACT-based immunotherapy stems from the activation of re-infused lymphocytes against a healthy tissue that expresses tumor-associated antigenic determinants, resulting in potentially lethal autoimmune reactions.Citation6,Citation10,Citation100,Citation101 This is relatively more frequent when ACT relies on PBLs that are genetically instructed to recognize malignant cells, possibly reflecting the capacity of exogenous, high-affinity TCRs or CARs to break immunological tolerance.Citation102,Citation103 Thus, the specificity of adoptively transferred cells determines not only the efficacy of ACT-based immunotherapeutic regimens but also their safety.Citation6 We have presented in previous Trial Watches how TAAs can be classified based on expression pattern and what are the advantages/disadvantages associated with the targeting of TAAs belonging to different of such categories.Citation104,Citation105

In previous issues of OncoImmunology, we detailed the scientific rationale behind the use of ACT for oncological indications and discussed recent clinical trials evaluating the immunotherapeutic profile of this regimen.Citation75,Citation76 Here, we present the latest advances in the development of ACT-based anticancer immunotherapy.

Literature Update

Since the submission of our previous Trial Watch dealing with topic (March 2013),Citation75 the results of no less than 14 clinical trials testing ACT-based immunotherapy in cancer patients have been published in the peer-reviewed scientific literature (source http://www.ncbi.nlm.nih.gov/pubmed). These studies involved patients affected by a relatively heterogeneous panel of hematological and solid malignancies, including acute lymphoblastic leukemia (ALL),Citation58,Citation106 acute myeloid leukemia (AML),Citation107 B-cell neoplasms,Citation108,Citation109 lymphoma,Citation110 synovial sarcoma,Citation101 melanoma,Citation101,Citation111,Citation112 as well as breast,Citation113 esophageal,Citation101 nasopharyngeal,Citation114 hepatocellular,Citation115 and renal carcinoma ().Citation116,Citation117

Table 1. Recently published clinical trials investigating the safety and efficacy of ACT-based immunotherapy in cancer patients

Brentjens and colleagues investigated the safety and efficacy of autologous T cells engineered to express a CD19-targeting CAR in 5 individuals with relapsed B-cell ALL.Citation58 All these patients achieved minimal residual disease-negative complete remission upon treatment. Moreover, this immunotherapeutic approach was generally well tolerated, although circulating cytokines raised significantly in some patients, a situation that had to be managed with lymphotoxic steroid therapy. Interestingly, the severity of cytokine elevations positively correlated with disease burden prior to therapy. This suggests that the cytokine-release syndromes that often develop in the course of ACT-based immunotherapy may predict clinical efficacy, at least in some scenarios.Citation58

Along similar lines, Grupp and coworkers reported on the clinical activity of autologous T lymphocytes engineered to express a CD19-targeting CAR in 2 children with relapsed and refractory pre-B-cell ALL.Citation106 In both cases, adoptively transferred T cells underwent a > 1000-fold expansion in vivo and colonized the bone marrow as well as the cerebrospinal fluid. A high number of Grade 3–4 side effects were recorded and both children developed a cytokine-release syndrome (which required pharmacological management in one case) and B-cell aplasia. This said, both patients underwent complete remission, lasting for more than 11 mo in one of them. The other patient relapsed 2 mo after treatment, owing to the emergence of a CD19- leukemic clone.Citation106 These observations lend further support to the notion that immunotherapeutic strategies targeting multiple TAAs may not only improve the safety of this approach but also its efficacy.Citation118,Citation119

Ritchie et al. investigated the safety and efficacy of autologous T cells redirected against the Lewis Y antigen (a difucosylated Type 2 blood group-related antigen)Citation120 in 4 AML patients previously subjected to fludarabine-based pre-conditioning.Citation107 No Grade 3–4 side effects were recorded and 3 patients experiences objective responses (including a protracted remission). Of note, CAR-expressing T cells were shown to actively traffic to the bone and to other proven sites of disease in the individuals who obtained the greatest benefits from treatment. Moreover, serial PCR-based analyses of the bone marrow and peripheral blood demonstrated that adoptively transferred T cells persisted for up to 10 mo in these patients.Citation107 This study demonstrates the safety and potential utility of autologous T cells retargeted against the Lewis Y antigen for the treatment of AML.

The group lead by Steven Rosenberg at the National Cancer Institute (Bethesda, MD USA) tested an innovative immunotherapeutic paradigm at the frontier between allogeneic HSCT and ACT in subjects with B-cell neoplasms that failed to respond to conventional allogeneic HSCT and donor lymphocyte infusions.Citation108 In this setting, 10 patients received a single infusion of allogeneic, rather than autologous, T lymphocytes engineered to express a CD19-specific CAR, in the absence of pre-conditioning. These cells were derived from the PBLs of each patient’s allogeneic HSCT donor. None of the individuals enrolled in this study developed severe toxicities including graft-vs.-host reactions, the most common side effects being transient hypotension and fever. However, 3 of them achieved objective responses, including a complete and a partial remission that were ongoing several months after therapy.Citation108

Cruz and collaborators investigated a similar approach in 8 patients with B-cell malignancies at high risk for relapse or relapsing upon allogeneic HSCT.Citation109 In this setting, however, donor T lymphocytes were first stimulated with autologous lymphoblastoid cell lines expressing viral antigens and then redirected against CD19 by the CAR technology. Indeed, virus-specific T cells (VSCs) infused into patients in the context of allogeneic HSCT are known to expand to significant levels, persist for long periods in vivo, and exhibit antiviral activity, but generally are unable to cause inducing graft-vs.-host reactions.Citation121,Citation122 Thus, Cruz and coworkers aimed at determining whether VSCs could be endowed with antitumor activity while preserving these positive features. No infusion-related toxicities were documented and 2 patients with relapsing disease experienced objective responses to treatment. Moreover, 2 patients who were in remission at infusion remained so 8 wk and 8 mo thereafter.Citation109 Together with the findings by Kochenderfer et al. discussed above,Citation108 these observations suggest that not only autologous, but also allogeneic, T cells may be genetically engineered to target specifically TAAs. The actual advantages of this approach remain to be determined.

Bollard and colleagues tested the therapeutic potential of autologous Epstein–Barr virus (EBV)-specific T lymphocytes expanded from EBV-related lymphoma patients by means of autologous DCs and EBV-transformed B-lymphoblastoid cell lines engineered to express EBV proteins.Citation110 Fifty patients were included in this study, none of whom manifested ACT-related side effects. Among 29 individuals affected by high-risk or previously relapsing disease at the time of infusion, 28 were in remission for a median time of 3.1 y, while 13 of the remaining 21 patients exhibited an objective response (including 11 complete responses). Of note, while T lymphocytes specific for EBV-encoded proteins were documented in the peripheral blood of a majority of patients, T cells reacting against non-viral TAAs (an indication of epitope spreading)Citation123,Citation124 could be detected only in individuals achieving clinical responses.Citation110 It remains unclear whether epitope spreading actually determines or is simply associated with the clinical activity of ACT-based immunotherapy in this setting.

Dudley and coworkers prospectively assigned 69 subjects with metastatic melanoma to receive adoptively transferred CD8+ T cell-enriched TILs (35 patients) or unselected “young” TILs (34 patients).Citation111 Young TILs have indeed been suggested to constitute a valuable alternative to their conventional counterparts, mainly because they are maintained in culture for a minimal amount of time and their preparation does not require individualized tumor-reactivity screening steps.Citation125-Citation127 Importantly, Dudley and colleagues demonstrated that CD8+ T cell-enriched TILs are not therapeutically superior to young TILs, at least for the treatment of advanced melanoma.Citation5,Citation111 This is relevant as the latter are not only easier to obtain than the former, but also significantly less expensive.Citation5

Besser et al. set out to investigate the therapeutic potential of young TILs combined with high-dose IL-2 in 80 Stage IV melanoma patients previously subjected to lymphodepleting pre-conditioning.Citation112 TIL cultures could be established from 72 out of 80 individuals, yet 15 of these subjects were withdrawn from the study owing to clinical deterioration prior to infusion. Of 57 patients receiving ACT-based immunotherapy, 18 achieved partial and 5 complete remission, the latter persisting in the absence of additional interventions 29 mo after treatment. Several factors were identified as independent predictors of clinical responses, including age, gender, circulating lactate dehydrogenase levels, days of TILs in culture, and number of infused cells.Citation112 Interestingly, 32 patients received ipilimumab, a FDA-approved monoclonal antibody specific for cytotoxic T lymphocyte assocaited protein 4 (CTLA4),Citation128-Citation130 prior or after the adoptive transfer of young TILs. Retrospective analyses demonstrated that the failure of these patients to respond to ipilimumab did not alter their propensity to benefit from ACT-based immunotherapy.Citation112

The study conducted by Domschke and collaborators enrolled 16 metastatic breast cancer patients bearing tumor-reactive memory T cells in the bone marrow.Citation113 These subjects received 1 infusion of autologous tumor-reactive T cells exposed ex vivo to DCs pulsed with breast carcinoma MCF-7 cell lysates, and 7 of them manifested an immunological response to therapy. Of note, responsiveness to therapy positively correlated with (1) absence of bone metastases, (2) amount of tumor-reactive T cells in the bone marrow at baseline, and (3) estimated number of adoptively transferred cells. Moreover, the overall survival of immunological responders was significantly higher than that of non-responders.Citation113 Although this clinical study was not designed for (nor sufficiently powered to) detect the therapeutic activity of ACT-based immunotherapy, the results from Domschke and colleagues confirm the prognostic/predictive value of (at least some) immunological parameters in cancer therapy.

Chia and colleagues tested the adoptive transfer of EBV-specific cytotoxic T lymphocytes in combination with gemcitabine (a potentially immunogenic nucleoside analog)Citation99,Citation131 and carboplatin (a platinum derivative with limited immunogenic potential)Citation132,Citation133 as a first-line therapeutic intervention in 35 patients with metastatic and/or locally recurrent EBV+ nasopharyngeal carcinoma.Citation114 No patient experienced severe (Grade > 3) toxicities in the course of therapy, and the most common adverse effects were Grade 1–2 fatigue and Grade 1 myalgia. Importantly, such an ACT-based immunotherapeutic regimen yielded an overall response rate of 71.4% (encompassing 3 complete and 22 partial responses), median overall and disease-free survival being 29.9 and 7.6 mo, respectively.Citation114 The outcome data reported in this study are among the best ever recorded in the setting of advanced EBV+ nasopharyngeal carcinoma, encouraging the evaluation of ACT-based immunochemotherapy in various other clinical settings.

Shimizu et al. investigated the clinical profile of T lymphocytes activated ex vivo plus DCs pulsed with autologous tumor lysates as an adjuvant intervention in patients with invasive hepatocellular carcinoma (HCC).Citation115 In this setting, 42 individuals received ACT-based immunotherapy upon surgical tumor resection, whereas 52 subjects underwent surgery only. No Grade 3–4 toxicities were recorded, the most common side effect associated with adjuvant cell-based immunotherapy being Grade 1 skin reactions at the injection site. Moreover, adjuvant immunotherapy yielded 5-y overall and disease-free survival rates of 64.3% and 35.7%, respectively, which compared favorably with those related to surgical resection alone (44.2% and 11.5%, respectively).Citation115 Interestingly, resected HCC patients developing delayed-type hypersensitivity (DTH) upon cell-based immunotherapy had a better prognosis than those who failed to do so.Citation115 This finding is in line previous results indicating that DTH reactions may predict the response of cancer patients to various immunotherapeutic regimens.Citation134-Citation136

Wang and colleagues evaluated the safety and therapeutic potential of cytokine-induced killer (CIK) cells combined with high-dose IL-2 in 29 patients with metastatic RCC.Citation116 CIK cells are a heterogeneous mix of effector CD8+ T cells that exerts MHC-unrestricted cytotoxic activity against malignant cells.Citation137-Citation139 The immunotherapeutic regimen designed by Wang and collaborators failed to induce complete responses, yet yielded a partial response in 4 individuals (13.8%) and disease stabilization in 18 subjects (62.1%). The 1-y overall survival rate was 82.8%. Of note, an inverse correlation was noted between the circulating levels of MDSCs at baseline and the propensity of RCC patients to obtain a survival benefit from therapy.Citation116 These findings not only suggest that the amounts of circulating MDSCs may predict the response of patients to ACT-based immunotherapy, but also that pharmacological interventions that deplete MDSCs, such as the administration of metronomic gemcitabine,Citation140,Citation141 may potentiate the clinical activity of adoptively transferred lymphocytes.

Lamers and coworkers enrolled 12 patients bearing carbonic anhydrase IX (CA9)-expressing RCC in a clinical study evaluating the activity of autologous T lymphocytes expressing a CA9-targeting CAR.Citation117,Citation142 These patients received a maximum of 10 infusions containing 0.2–2.1 x 109 T lymphocytes. Grade 2–4 elevations of circulating hepatic enzymes were recorded in 8 patients treated with the lowest T-cell dose. In line with these findings, T cells were found to infiltrate the bile duct epithelium in bioptic specimens, correlating with the local expression of CA9.Citation117 Of note, the pre-administration of CA9-targeting monoclonal antibodies prevented such toxicities, pointing to a strategy for avoiding (or at least limiting) the on-target side effects of ACT-based immunotherapy.

Morgan et al. evaluated the safety and therapeutic potential of autologous T lymphocytes engineered to express a melanoma antigen family A3 (MAGEA3)-specific TCR in 9 patients affected by MAGEA3+ malignancies, including melanoma, synovial sarcoma, and esophageal carcinoma.Citation6,Citation101 While 5 patients experienced tumor regression, 3 developed severe mental status changes beginning 1–2 post-infusion. In one of such patients, initial Parkinson-like symptoms resolved over 4 wk. Conversely, in the 2 other cases, the situation rapidly degenerated into coma and death. Autoptic studies revealed a necrotizing leukoencephalopathy with extensive white matter defects associated with the infiltration of CD3+/CD8+ T cells.Citation6,Citation101 Prompted by these findings, Morgan and colleagues investigated in detail the expression profile of MAGEA family members in the human brain, identifying in MAGE-A12 the most solid candidate for the unexpected toxicity of their ACT-based immunotherapeutic protocol.Citation101

Among recent translational studies investigating ACT-based immunotherapy, we found of particular interest the works of (1) Alizadeh and colleagues, who demonstrated that doxorubicin (an immunogenic anthracycline)Citation143-Citation145 may improve the therapeutic potential of adoptively transferred lymphocytes as it depletes MDSCs, in thus far resembling gemcitabine; (2) Budde et al., who endowed T cells expressing a CD20-specific CAR with an inducible suicide system based on the pharmacological dimerization of caspase-9, allowing for their rapid elimination in vitro and in vivo;Citation146 (3) Labarriere and collaborators, who developed and validated a full good manufacturing practice (GMP) process to select and amplify melan A (MLANA) and MELOE-1-specific T cells for the treatment of metastatic melanoma patients from 100 mL of peripheral blood;Citation147 (4) Ma and coworkers, who studied the functional activity and dynamics of T lymphocytes expressing a MLANA-specific TCR upon reinfusion in melanoma patients;Citation148 (5) Karlsson et al., who demonstrated that the cytotoxic activity of T lymphocytes bearing a CD19-specific CAR can be boosted by the co- or pre-administration of ABT-737, a small molecule inhibitor of anti-apoptotic BCL-2 family members,Citation149-Citation152 at least in vitro;Citation153 (6) Mardiros and colleagues, who developed and demonstrated the specificity as well as therapeutic potential of 2 distinct CARs specific for IL-3 receptor, α (IL3RA, best known as CD123);Citation154 and (7) Robbins and coworkers, who designed and validated a whole exome sequencing-based screening procedure to identify neo-antigens potentially recognized by TILs.Citation155 This approach may be of particular translational value as it may allow for the relatively straightforward selection and expansion of TILs that recognize patient-specific TAAs.

Taken together, these observations indicate that several groups worldwide firmly believe in the clinical potential of ACT-based immunotherapy and are investing consistent efforts in the development of safe, efficient and cost-effective ACT protocols.

Update on Ongoing Clinical Trials

When this Trial Watch was being redacted (February 2014), official sources listed no less than 33 clinical trials launched after March 1st, 2013 that would evaluate the efficacy and safety of ACT-based immunotherapy in cancer patients (source http://www.clinicaltrials.gov).

In line with an increasing amount of encouraging clinical findings,Citation48,Citation49,Citation56,Citation58,Citation106 a significant fraction of the clinical trials launched in the last 12 mo involves the administration CAR-expressing T cells to individuals with hematological neoplasms, notably ALL, chronic lymphocytic leukemia, and non-Hodgkin lymphoma (). With a few exceptions (NCT01815749; NCT01840566), these studies rely on ACT-based immunotherapy as a standalone intervention in the context of lymphodepleting pre-conditioning (NCT01853631; NCT01860937; NCT01864889; NCT01864902; NCT01865617; NCT02028455; NCT02030847; NCT02050347; NCT02051257). Conversely, in NCT01815749 and NCT01840566, genetically modified autologous T cells are administered to patients upon myeloablative pre-conditioning and allogeneic or autologous HSCT. In addition, ACT-based immunotherapy is being tested in various cohorts of multiple myeloma patients. These clinical trials involve (1) autologous HSCT followed by the infusion of activated marrow-infiltrating lymphocytes and lenalidomide-based maintenance therapyCitation156-Citation159 (NCT01858558); (2) the administration of autologous T cells manipulated to express a TCR specific for the cancer/testis antigens NY-ESO-1 and cancer/testis antigen 2 (CTA2, best known as LAGE-1)Citation160-Citation162 (NCT01892293); and (3) the infusion of autologous or donor-derived T cells stably transduced to express a CAR targeting syndecan 1 (SDC1, best known as CD138)Citation163,Citation164 (NCT01886976).

Table 2. Clinical trials recently launched to evaluate the safety and efficacy of ACT-based immunotherapy in cancer patients.*

Reflecting the particular sensitivity of melanoma to immunotherapeutic interventions,Citation16,Citation165-Citation167 no less than 9 clinical trials have been initiated during the last 12 mo to investigate the safety and therapeutic potential of ACT-based regimens in this oncological setting (). Most of these studies involve the administration to pre-conditioned patients of TILs expanded ex vivo following conventional procedures, in combination with high- or low-dose IL-2 (NCT01807182; NCT01814046; NCT01883323; NCT01946373; NCT01995344). Alternatively, (1) TILs are infused together with low-dose IL-2 in the absence of pre-conditioning (NCT01883297); (2) TILs are administered to pre-conditioned patients in combination with DCs pulsed ex vivo with autologous tumor lysates and a NY-ESO-1-derived peptide (NCT01946373); (3) young TILs are co-administered with IL-2 in a trial comparing 2 distinct pre-conditioning regimens (NCT01993719); or (4) PBLs engineered to express a dominant negative variant of the transforming growth factor β1 (TGFβ1) receptor are administered to pre-conditioned patients, followed by high-dose IL-2 (NCT01955460). Finally, 1 study aims at assessing the clinical profile of autologous CD8+ PBLs given in combination with ipilimumab (NCT02027935).

Hematological cancers and melanoma are not the sole oncological indications in which ACT-based immunotherapy is being actively investigated (). Indeed, during the last 12 mo several clinical trials have been initiated to test: (1) T lymphocytes expressing a ganglioside D2-specific CAR and endowed with an inducible suicide system, in neuroblastoma patients (NCT01822652); (2) activated T lymphocytes combined with the multi-kinase inhibitor sorafenib,Citation168-Citation171 in HCC patients; (3) CIK cells as standalone therapeutic intervention, in subjects affected by cholangiocarcinoma (NCT01868490) or colorectal carcinoma (NCT01839539); (4) autologous T cells manipulated to express a CAR specific for v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2, best known as HER2),Citation172,Citation173 in patients bearing HER2+ solid neoplasms (NCT01935843); (5) autologous T lymphocytes expressing a CAR specific for epidermal growth factor receptor (EGFR),Citation174,Citation175 in subjects with advanced EGFR+ malignancies (NCT01869166); and (6) autologous PBLs genetically modified to express a NY-ESO-1-targeting TCR, in subjects affected by metastatic NY-ESO-1+ tumors (NCT01967823).

As for the clinical trials listed in our previous Trial Watches dealing with this topic,Citation75,Citation76 the following studies have changed status during the past 12 mo: NCT01236573, now listed as “Suspended”; NCT00871481, now listed as “Completed”; as well as NCT01555892, NCT01567891, NCT01653717, NCT01729091, and NCT01758458, now listed as “Recruiting.” Nor the reasons underlying the suspension of NCT01236573, testing IL-12-expressing TILs combined with ipilimumab in metastatic melanoma patients, nor the results of NCT00871481, investigating the therapeutic profile of NY-ESO-1-redirected TILs combined with IL-2 and ipilimumab in a similar setting, appear to be available (source http://www.clinicaltrials.gov).

In summary, the enthusiasm that has gathered around ACT-based immunotherapy throughout the past 2 decades remains high. The use of autologous CAR-expressing T lymphocytes for the treatment of B-cell neoplasms or genetically unmodified TILs for the therapy of melanoma stand out as the protocols of this type most intensively evaluated in the clinics today.

Concluding Remarks

Accumulating evidence suggests that ACT-based anticancer immunotherapy may soon cease to be a promising experimental regimen and become an established clinical reality. This said, the 2 treatment-related deaths recorded by Morgan and colleagues upon the infusion of autologous T cells expressing a MAGE-A3-targeting CARCitation6,Citation101 should warn the scientific and medical community about the potential downsides of this approach.

As for many other TAA-specific immunotherapeutic regimens, including DNA-based as well as peptide-based vaccines,Citation176,Citation177 the efficacy as well as the safety of ACT-based interventions relying on genetically engineered T cells depends on the TAA of choice.Citation178 Strategies based on the simultaneous targeting of 2 distinct TAAs are being actively investigated, not only as they would be associated with limited on-target toxicity for normal tissues that express one single TAA, but also as they may limit the surge of antigen-loss tumor variants.Citation119,Citation179,Citation180 Moreover, the safety and efficacy of ACT-based immunotherapeutic regimens relying on genetically manipulated T lymphocytes appear to depend on the affinity and avidity of exogenously introduced TAA receptors, be them TCRs or CARs.Citation102,Citation103,Citation181 Significant efforts have been dedicated at the development of 2nd and 3rd generation CARs that deliver potent immunostimulatory cues to T cells, de facto bypassing the need for co-stimulatory signaling.Citation182,Citation183 The recent unfortunate death of 2 patients treated with CD123-redirected T cells suggests that the successful clinical application of some of these CARs requires adequate brakes.

Abbreviations:
ACT=

adoptive cell transfer

ALL=

acute lymphoblastic leukemia

AML=

acute myeloid leukemia

CA9=

carbonic anhydrase IX

CAR=

chimeric antigen receptor

CIK=

cytokine-induced killer

DC=

dendritic cell

DTH=

delayed-type hypersensitivity

EBV=

Epstein-Barr virus

EGFR=

epidermal growth factor receptor

FDA=

Food and Drug Administration

HCC=

hepatocellular carcinoma

HSCT=

hematopoietic stem cell transplantation

IL=

interleukin

MAGEA3=

melanoma antigen family A3

MDSC=

myeloid-derived suppressor cell

MLANA=

melan A

PBL=

peripheral blood lymphocyte

RCC=

renal cell carcinoma

TAA=

tumor-associated antigen

TCR=

T-cell receptor

TIL=

tumor-infiltrating lymphocyte

VSC=

virus-specific T cell

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).

Citation: Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Hervé Fridman W, Cremer I, Tartour E, Zitvogel L, Kroemer G, et al. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. OncoImmunology 2014; 3:e28344; 10.4161/onci.28344

References

  • Speiser DE. Hit parade for adoptive cell transfer therapy: the best T cells for superior clinical responses. Cancer Discov 2013; 3:379 - 81; http://dx.doi.org/10.1158/2159-8290.CD-13-0064; PMID: 23580281
  • Yee C. Adoptive T-cell therapy for cancer: boutique therapy or treatment modality?. Clin Cancer Res 2013; 19:4550 - 2; http://dx.doi.org/10.1158/1078-0432.CCR-13-1367; PMID: 23922301
  • Ruella M, Kalos M. Adoptive immunotherapy for cancer. Immunol Rev 2014; 257:14 - 38; http://dx.doi.org/10.1111/imr.12136; PMID: 24329787
  • Yee C. The use of endogenous T cells for adoptive transfer. Immunol Rev 2014; 257:250 - 63; http://dx.doi.org/10.1111/imr.12134; PMID: 24329802
  • Kirk R. Immunotherapy: Adoptive cell therapy simplified. Nat Rev Clin Oncol 2013; 10:368; http://dx.doi.org/10.1038/nrclinonc.2013.85; PMID: 23689751
  • Humphries C. Adoptive cell therapy: Honing that killer instinct. Nature 2013; 504:S13 - 5; http://dx.doi.org/10.1038/504S13a; PMID: 24352359
  • Maus MV, Fraietta JA, Levine BL, Kalos M, Zhao Y, June CH. Adoptive Immunotherapy for Cancer or Viruses. Annu Rev Immunol 2014; Forthcoming PMID: 24423116
  • Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol 2012; 12:269 - 81; http://dx.doi.org/10.1038/nri3191; PMID: 22437939
  • Morgan RA, Dudley ME, Rosenberg SA. Adoptive cell therapy: genetic modification to redirect effector cell specificity. Cancer J 2010; 16:336 - 41; http://dx.doi.org/10.1097/PPO.0b013e3181eb3879; PMID: 20693844
  • Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008; 8:299 - 308; http://dx.doi.org/10.1038/nrc2355; PMID: 18354418
  • 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; http://dx.doi.org/10.4161/onci.21494; PMID: 23170259
  • 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; http://dx.doi.org/10.4161/onci.25771; PMID: 24286020
  • Ueno H, Klechevsky E, Schmitt N, Ni L, Flamar AL, Zurawski S, Zurawski G, Palucka K, Banchereau J, Oh S. Targeting human dendritic cell subsets for improved vaccines. Semin Immunol 2011; 23:21 - 7; http://dx.doi.org/10.1016/j.smim.2011.01.004; PMID: 21277223
  • Ueno H, Palucka AK, Banchereau J. The expanding family of dendritic cell subsets. Nat Biotechnol 2010; 28:813 - 5; http://dx.doi.org/10.1038/nbt0810-813; PMID: 20697407
  • Palucka K, Banchereau J, Mellman I. Designing vaccines based on biology of human dendritic cell subsets. Immunity 2010; 33:464 - 78; http://dx.doi.org/10.1016/j.immuni.2010.10.007; PMID: 21029958
  • Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, 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; 23:2346 - 57; http://dx.doi.org/10.1200/JCO.2005.00.240; PMID: 15800326
  • Jenq RR, van den Brink MR. Allogeneic haematopoietic stem cell transplantation: individualized stem cell and immune therapy of cancer. Nat Rev Cancer 2010; 10:213 - 21; http://dx.doi.org/10.1038/nrc2804; PMID: 20168320
  • Barriga F, Ramírez P, Wietstruck A, Rojas N. Hematopoietic stem cell transplantation: clinical use and perspectives. Biol Res 2012; 45:307 - 16; http://dx.doi.org/10.4067/S0716-97602012000300012; PMID: 23283440
  • Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, Jungbluth A, Gnjatic S, Thompson JA, Yee C. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med 2008; 358:2698 - 703; http://dx.doi.org/10.1056/NEJMoa0800251; PMID: 18565862
  • Pegram HJ, Jackson JT, Smyth MJ, Kershaw MH, Darcy PK. Adoptive transfer of gene-modified primary NK cells can specifically inhibit tumor progression in vivo. J Immunol 2008; 181:3449 - 55; PMID: 18714017
  • Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, Posati S, Rogaia D, Frassoni F, Aversa F, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002; 295:2097 - 100; http://dx.doi.org/10.1126/science.1068440; PMID: 11896281
  • Velardi A, Ruggeri L, Mancusi A, Aversa F, Christiansen FT. Natural killer cell allorecognition of missing self in allogeneic hematopoietic transplantation: a tool for immunotherapy of leukemia. Curr Opin Immunol 2009; 21:525 - 30; http://dx.doi.org/10.1016/j.coi.2009.07.015; PMID: 19717293
  • Ohira M, Ohdan H, Mitsuta H, Ishiyama K, Tanaka Y, Igarashi Y, Asahara T. Adoptive transfer of TRAIL-expressing natural killer cells prevents recurrence of hepatocellular carcinoma after partial hepatectomy. Transplantation 2006; 82:1712 - 9; http://dx.doi.org/10.1097/01.tp.0000250935.41034.2d; PMID: 17198265
  • Okada K, Nannmark U, Vujanovic NL, Watkins S, Basse P, Herberman RB, Whiteside TL. Elimination of established liver metastases by human interleukin 2-activated natural killer cells after locoregional or systemic adoptive transfer. Cancer Res 1996; 56:1599 - 608; PMID: 8603408
  • Besser MJ, Shoham T, Harari-Steinberg O, Zabari N, Ortenberg R, Yakirevitch A, Nagler A, Loewenthal R, Schachter J, Markel G. Development of allogeneic NK cell adoptive transfer therapy in metastatic melanoma patients: in vitro preclinical optimization studies. PLoS One 2013; 8:e57922; http://dx.doi.org/10.1371/journal.pone.0057922; PMID: 23483943
  • Terme M, Fridman WH, Tartour E. NK cells from pleural effusions are potent antitumor effector cells. Eur J Immunol 2013; 43:331 - 4; http://dx.doi.org/10.1002/eji.201243264; PMID: 23322344
  • Lister J, Rybka WB, Donnenberg AD, deMagalhaes-Silverman M, Pincus SM, Bloom EJ, Elder EM, Ball ED, Whiteside TL. Autologous peripheral blood stem cell transplantation and adoptive immunotherapy with activated natural killer cells in the immediate posttransplant period. Clin Cancer Res 1995; 1:607 - 14; PMID: 9816022
  • Parkhurst MR, Riley JP, Dudley ME, Rosenberg SA. Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin Cancer Res 2011; 17:6287 - 97; http://dx.doi.org/10.1158/1078-0432.CCR-11-1347; PMID: 21844012
  • Iliopoulou EG, Kountourakis P, Karamouzis MV, Doufexis D, Ardavanis A, Baxevanis CN, Rigatos G, Papamichail M, Perez SA. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother 2010; 59:1781 - 9; http://dx.doi.org/10.1007/s00262-010-0904-3; PMID: 20703455
  • Badoual C, Bastier P-L, Roussel H, Mandavit M, Tartour E. An allogeneic NK cell line engineered to express chimeric antigen receptors: A novel strategy of cellular immunotherapy against cancer. OncoImmunology 2013; 2:e27156; http://dx.doi.org/10.4161/onci.27156
  • Boissel L, Betancur-Boissel M, Lu W, Krause DS, Van Etten RA, Wels WS, Klingemann H. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology 2013; 2:e26527; http://dx.doi.org/10.4161/onci.26527; PMID: 24404423
  • Altvater B, Landmeier S, Pscherer S, Temme J, Schweer K, Kailayangiri S, Campana D, Juergens H, Pule M, Rossig C. 2B4 (CD244) signaling by recombinant antigen-specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells. Clin Cancer Res 2009; 15:4857 - 66; http://dx.doi.org/10.1158/1078-0432.CCR-08-2810; PMID: 19638467
  • Boissel L, Betancur M, Wels WS, Tuncer H, Klingemann H. Transfection with mRNA for CD19 specific chimeric antigen receptor restores NK cell mediated killing of CLL cells. Leuk Res 2009; 33:1255 - 9; http://dx.doi.org/10.1016/j.leukres.2008.11.024; PMID: 19147228
  • Li Q, Lao X, Pan Q, Ning N, Yet J, Xu Y, Li S, Chang AE. Adoptive transfer of tumor reactive B cells confers host T-cell immunity and tumor regression. Clin Cancer Res 2011; 17:4987 - 95; http://dx.doi.org/10.1158/1078-0432.CCR-11-0207; PMID: 21690573
  • de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell 2005; 7:411 - 23; http://dx.doi.org/10.1016/j.ccr.2005.04.014; PMID: 15894262
  • Schioppa T, Moore R, Thompson RG, Rosser EC, Kulbe H, Nedospasov S, Mauri C, Coussens LM, Balkwill FR. B regulatory cells and the tumor-promoting actions of TNF-α during squamous carcinogenesis. Proc Natl Acad Sci U S A 2011; 108:10662 - 7; http://dx.doi.org/10.1073/pnas.1100994108; PMID: 21670304
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2012; 1:1323 - 43; http://dx.doi.org/10.4161/onci.22009; PMID: 23243596
  • Yun YS, Hargrove ME, Ting CC. In vivo antitumor activity of anti-CD3-induced activated killer cells. Cancer Res 1989; 49:4770 - 4; PMID: 2527087
  • Bouquié R, Bonnin A, Bernardeau K, Khammari A, Dréno B, Jotereau F, Labarrière N, Lang F. A fast and efficient HLA multimer-based sorting procedure that induces little apoptosis to isolate clinical grade human tumor specific T lymphocytes. Cancer Immunol Immunother 2009; 58:553 - 66; http://dx.doi.org/10.1007/s00262-008-0578-2; PMID: 18751701
  • Rosenberg SA. Cell transfer immunotherapy for metastatic solid cancer--what clinicians need to know. Nat Rev Clin Oncol 2011; 8:577 - 85; http://dx.doi.org/10.1038/nrclinonc.2011.116; PMID: 21808266
  • Chhabra A, Yang L, Wang P, Comin-Anduix B, Das R, Chakraborty NG, Ray S, Mehrotra S, Yang H, Hardee CL, et al. CD4+CD25- T cells transduced to express MHC class I-restricted epitope-specific TCR synthesize Th1 cytokines and exhibit MHC class I-restricted cytolytic effector function in a human melanoma model. J Immunol 2008; 181:1063 - 70; PMID: 18606658
  • Ray S, Chhabra A, Chakraborty NG, Hegde U, Dorsky DI, Chodon T, von Euw E, Comin-Anduix B, Koya RC, Ribas A, et al, UCLA-CALTECH-CHLA-USC-UCONN Consortium on Translational Program in Engineered Immunity. MHC-I-restricted melanoma antigen specific TCR-engineered human CD4+ T cells exhibit multifunctional effector and helper responses, in vitro. Clin Immunol 2010; 136:338 - 47; http://dx.doi.org/10.1016/j.clim.2010.04.013; PMID: 20547105
  • Sadelain M, Rivière I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer 2003; 3:35 - 45; http://dx.doi.org/10.1038/nrc971; PMID: 12509765
  • Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV, Helman LJ, Mackall CL, 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; 29:917 - 24; http://dx.doi.org/10.1200/JCO.2010.32.2537; PMID: 21282551
  • Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discov 2013; 3:388 - 98; http://dx.doi.org/10.1158/2159-8290.CD-12-0548; PMID: 23550147
  • Dotti G, Gottschalk S, Savoldo B, Brenner MK. Design and development of therapies using chimeric antigen receptor-expressing T cells. Immunol Rev 2014; 257:107 - 26; http://dx.doi.org/10.1111/imr.12131; PMID: 24329793
  • Jensen MC, Riddell SR. Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev 2014; 257:127 - 44; http://dx.doi.org/10.1111/imr.12139; PMID: 24329794
  • Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365:725 - 33; http://dx.doi.org/10.1056/NEJMoa1103849; PMID: 21830940
  • Kochenderfer JN, Rosenberg SA. Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat Rev Clin Oncol 2013; 10:267 - 76; http://dx.doi.org/10.1038/nrclinonc.2013.46; PMID: 23546520
  • Long AH, Haso WM, Orentas RJ. Lessons learned from a highly-active CD22-specific chimeric antigen receptor. Oncoimmunology 2013; 2:e23621; http://dx.doi.org/10.4161/onci.23621; PMID: 23734316
  • Spear P, Barber A, Sentman CL. Collaboration of chimeric antigen receptor (CAR)-expressing T cells and host T cells for optimal elimination of established ovarian tumors. Oncoimmunology 2013; 2:e23564; http://dx.doi.org/10.4161/onci.23564; PMID: 23734311
  • Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M, Phan GQ, Hughes MS, Sherry RM, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012; 119:2709 - 20; http://dx.doi.org/10.1182/blood-2011-10-384388; PMID: 22160384
  • Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I, Raffeld M, Nathan DA, Lanier BJ, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010; 116:4099 - 102; http://dx.doi.org/10.1182/blood-2010-04-281931; PMID: 20668228
  • Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 2010; 116:3875 - 86; http://dx.doi.org/10.1182/blood-2010-01-265041; PMID: 20631379
  • Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, Taylor C, Yeh R, Bartido S, Borquez-Ojeda O, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011; 118:4817 - 28; http://dx.doi.org/10.1182/blood-2011-04-348540; PMID: 21849486
  • Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 2011; 3:95ra73; http://dx.doi.org/10.1126/scitranslmed.3002842; PMID: 21832238
  • Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, Kamble RT, Bollard CM, Gee AP, Mei Z, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 2011; 121:1822 - 6; http://dx.doi.org/10.1172/JCI46110; PMID: 21540550
  • Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 2013; 5:177ra38; http://dx.doi.org/10.1126/scitranslmed.3005930; PMID: 23515080
  • Merhavi-Shoham E, Haga-Friedman A, Cohen CJ. Genetically modulating T-cell function to target cancer. Semin Cancer Biol 2012; 22:14 - 22; http://dx.doi.org/10.1016/j.semcancer.2011.12.006; PMID: 22210183
  • Liu K, Rosenberg SA. Transduction of an IL-2 gene into human melanoma-reactive lymphocytes results in their continued growth in the absence of exogenous IL-2 and maintenance of specific antitumor activity. J Immunol 2001; 167:6356 - 65; PMID: 11714800
  • Zhou J, Shen X, Huang J, Hodes RJ, Rosenberg SA, Robbins PF. Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy. J Immunol 2005; 175:7046 - 52; PMID: 16272366
  • Kalbasi A, Shrimali RK, Chinnasamy D, Rosenberg SA. Prevention of interleukin-2 withdrawal-induced apoptosis in lymphocytes retrovirally cotransduced with genes encoding an antitumor T-cell receptor and an antiapoptotic protein. J Immunother 2010; 33:672 - 83; http://dx.doi.org/10.1097/CJI.0b013e3181e475cd; PMID: 20664359
  • Hinrichs CS, Borman ZA, Gattinoni L, Yu Z, Burns WR, Huang J, Klebanoff CA, Johnson LA, Kerkar SP, Yang S, et al. Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood 2011; 117:808 - 14; http://dx.doi.org/10.1182/blood-2010-05-286286; PMID: 20971955
  • Bellone M, Calcinotto A, Corti A. Won’t you come on in? How to favor lymphocyte infiltration in tumors. Oncoimmunology 2012; 1:986 - 8; http://dx.doi.org/10.4161/onci.20213; PMID: 23162781
  • Kershaw MH, Teng MW, Smyth MJ, Darcy PK. Supernatural T cells: genetic modification of T cells for cancer therapy. Nat Rev Immunol 2005; 5:928 - 40; http://dx.doi.org/10.1038/nri1729; PMID: 16322746
  • Galluzzi L, Lugli E. Rejuvenated T cells attack old tumors. Oncoimmunology 2013; 2:e24103; http://dx.doi.org/10.4161/onci.24103; PMID: 23526137
  • Gattinoni L, Klebanoff CA, Restifo NP. Paths to stemness: building the ultimate antitumour T cell. Nat Rev Cancer 2012; 12:671 - 84; http://dx.doi.org/10.1038/nrc3322; PMID: 22996603
  • Somerville RP, Dudley ME. Bioreactors get personal. Oncoimmunology 2012; 1:1435 - 7; http://dx.doi.org/10.4161/onci.21206; PMID: 23243620
  • Cieri N, Camisa B, Cocchiarella F, Forcato M, Oliveira G, Provasi E, Bondanza A, Bordignon C, Peccatori J, Ciceri F, et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2013; 121:573 - 84; http://dx.doi.org/10.1182/blood-2012-05-431718; PMID: 23160470
  • Lugli E, Dominguez MH, Gattinoni L, Chattopadhyay PK, Bolton DL, Song K, Klatt NR, Brenchley JM, Vaccari M, Gostick E, et al. Superior T memory stem cell persistence supports long-lived T cell memory. J Clin Invest 2013; 123:594 - 9; PMID: 23281401
  • Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, Almeida JR, Gostick E, Yu Z, Carpenito C, et al. A human memory T cell subset with stem cell-like properties. Nat Med 2011; 17:1290 - 7; http://dx.doi.org/10.1038/nm.2446; PMID: 21926977
  • Nishimura T, Kaneko S, Kawana-Tachikawa A, Tajima Y, Goto H, Zhu D, Nakayama-Hosoya K, Iriguchi S, Uemura Y, Shimizu T, et al. Generation of rejuvenated antigen-specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell 2013; 12:114 - 26; http://dx.doi.org/10.1016/j.stem.2012.11.002; PMID: 23290140
  • Haymaker C, Wu R, Bernatchez C, Radvanyi L. PD-1 and BTLA and CD8(+) T-cell “exhaustion” in cancer: “Exercising” an alternative viewpoint. Oncoimmunology 2012; 1:735 - 8; http://dx.doi.org/10.4161/onci.20823; PMID: 22934265
  • Vizcardo R, Masuda K, Yamada D, Ikawa T, Shimizu K, Fujii S, Koseki H, Kawamoto H. Regeneration of human tumor antigen-specific T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell 2013; 12:31 - 6; http://dx.doi.org/10.1016/j.stem.2012.12.006; PMID: 23290135
  • 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; http://dx.doi.org/10.4161/onci.24238; PMID: 23762803
  • 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; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • Wrzesinski C, Paulos CM, Kaiser A, Muranski P, Palmer DC, Gattinoni L, Yu Z, Rosenberg SA, Restifo NP. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother 2010; 33:1 - 7; http://dx.doi.org/10.1097/CJI.0b013e3181b88ffc; PMID: 19952961
  • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9:162 - 74; http://dx.doi.org/10.1038/nri2506; PMID: 19197294
  • Montero AJ, Diaz-Montero CM, Kyriakopoulos CE, Bronte V, Mandruzzato S. Myeloid-derived suppressor cells in cancer patients: a clinical perspective. J Immunother 2012; 35:107 - 15; http://dx.doi.org/10.1097/CJI.0b013e318242169f; PMID: 22306898
  • Nagaraj S, Gabrilovich DI. Myeloid-derived suppressor cells in human cancer. Cancer J 2010; 16:348 - 53; http://dx.doi.org/10.1097/PPO.0b013e3181eb3358; PMID: 20693846
  • Cheng G, Yu A, Malek TR. T-cell tolerance and the multi-functional role of IL-2R signaling in T-regulatory cells. Immunol Rev 2011; 241:63 - 76; http://dx.doi.org/10.1111/j.1600-065X.2011.01004.x; PMID: 21488890
  • Rudensky AY. Regulatory T cells and Foxp3. Immunol Rev 2011; 241:260 - 8; http://dx.doi.org/10.1111/j.1600-065X.2011.01018.x; PMID: 21488902
  • Yao X, Ahmadzadeh M, Lu YC, Liewehr DJ, Dudley ME, Liu F, Schrump DS, Steinberg SM, Rosenberg SA, Robbins PF. Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 2012; 119:5688 - 96; http://dx.doi.org/10.1182/blood-2011-10-386482; PMID: 22555974
  • Kodumudi KN, Weber A, Sarnaik AA, Pilon-Thomas S. Blockade of myeloid-derived suppressor cells after induction of lymphopenia improves adoptive T cell therapy in a murine model of melanoma. J Immunol 2012; 189:5147 - 54; http://dx.doi.org/10.4049/jimmunol.1200274; PMID: 23100512
  • Pere H, Tanchot C, Bayry J, Terme M, Taieb J, Badoual C, Adotevi O, Merillon N, Marcheteau E, Quillien VR, et al. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology 2012; 1:326 - 33; http://dx.doi.org/10.4161/onci.18852; PMID: 22737608
  • Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, Hwang LN, Yu Z, Wrzesinski C, Heimann DM, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005; 202:907 - 12; http://dx.doi.org/10.1084/jem.20050732; PMID: 16203864
  • Klebanoff CA, Khong HT, Antony PA, Palmer DC, Restifo NP. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol 2005; 26:111 - 7; http://dx.doi.org/10.1016/j.it.2004.12.003; PMID: 15668127
  • Mignot G, Ullrich E, Bonmort M, Ménard C, Apetoh L, Taieb J, Bosisio D, Sozzani S, Ferrantini M, Schmitz J, et al. The critical role of IL-15 in the antitumor effects mediated by the combination therapy imatinib and IL-2. J Immunol 2008; 180:6477 - 83; PMID: 18453565
  • Ullrich E, Bonmort M, Mignot G, Jacobs B, Bosisio D, Sozzani S, Jalil A, Louache F, Bulanova E, Geissman F, et al. Trans-presentation of IL-15 dictates IFN-producing killer dendritic cells effector functions. J Immunol 2008; 180:7887 - 97; PMID: 18523252
  • Liu DL, Håkansson CH, Seifert J. Immunotherapy in liver tumors: II. Intratumoral injection with activated tumor-infiltrating lymphocytes, intrasplenic administration of recombinant interleukin-2 and interferon alpha causes tumor regression and lysis. Cancer Lett 1994; 85:39 - 46; http://dx.doi.org/10.1016/0304-3835(94)90236-4; PMID: 7923100
  • Helms MW, Prescher JA, Cao YA, Schaffert S, Contag CH. IL-12 enhances efficacy and shortens enrichment time in cytokine-induced killer cell immunotherapy. Cancer Immunol Immunother 2010; 59:1325 - 34; http://dx.doi.org/10.1007/s00262-010-0860-y; PMID: 20532883
  • Dings RP, Vang KB, Castermans K, Popescu F, Zhang Y, Oude Egbrink MG, Mescher MF, Farrar MA, Griffioen AW, Mayo KH. Enhancement of T-cell-mediated antitumor response: angiostatic adjuvant to immunotherapy against cancer. Clin Cancer Res 2011; 17:3134 - 45; http://dx.doi.org/10.1158/1078-0432.CCR-10-2443; PMID: 21252159
  • Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res 2010; 70:6171 - 80; http://dx.doi.org/10.1158/0008-5472.CAN-10-0153; PMID: 20631075
  • 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; http://dx.doi.org/10.4161/onci.20696; PMID: 22934262
  • Paulos CM, Kaiser A, Wrzesinski C, Hinrichs CS, Cassard L, Boni A, Muranski P, Sanchez-Perez L, Palmer DC, Yu Z, et al. Toll-like receptors in tumor immunotherapy. Clin Cancer Res 2007; 13:5280 - 9; http://dx.doi.org/10.1158/1078-0432.CCR-07-1378; PMID: 17875756
  • 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; http://dx.doi.org/10.4161/onci.20931; PMID: 23162757
  • Yang Y, Huang CT, Huang X, Pardoll DM. Persistent Toll-like receptor signals are required for reversal of regulatory T cell-mediated CD8 tolerance. Nat Immunol 2004; 5:508 - 15; http://dx.doi.org/10.1038/ni1059; PMID: 15064759
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74 - 88; http://dx.doi.org/10.1016/j.immuni.2013.06.014; PMID: 23890065
  • Yeh S, Karne NK, Kerkar SP, Heller CK, Palmer DC, Johnson LA, Li Z, Bishop RJ, Wong WT, Sherry RM, et al. Ocular and systemic autoimmunity after successful tumor-infiltrating lymphocyte immunotherapy for recurrent, metastatic melanoma. Ophthalmology 2009; 116:981 - , e1; http://dx.doi.org/10.1016/j.ophtha.2008.12.004; PMID: 19410956
  • Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, Dudley ME, Feldman SA, Yang JC, Sherry RM, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 2013; 36:133 - 51; http://dx.doi.org/10.1097/CJI.0b013e3182829903; PMID: 23377668
  • Zhong S, Malecek K, Johnson LA, Yu Z, Vega-Saenz de Miera E, Darvishian F, McGary K, Huang K, Boyer J, Corse E, et al. T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci U S A 2013; 110:6973 - 8; http://dx.doi.org/10.1073/pnas.1221609110; PMID: 23576742
  • Wilde S, Schendel DJ. High-quality and high-avidity T cell clones specific for tumor-associated antigens and how to find them. Oncoimmunology 2012; 1:1643 - 4; http://dx.doi.org/10.4161/onci.21717; PMID: 23264922
  • Senovilla L, Vacchelli E, Garcia P, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: DNA vaccines for cancer therapy. Oncoimmunology 2013; 2:e23803; http://dx.doi.org/10.4161/onci.23803; PMID: 23734328
  • 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; http://dx.doi.org/10.4161/onci.22428; PMID: 23264902
  • Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013; 368:1509 - 18; http://dx.doi.org/10.1056/NEJMoa1215134; PMID: 23527958
  • Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K, Chen K, Shin M, Wall DM, Hönemann D, et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol Ther 2013; 21:2122 - 9; http://dx.doi.org/10.1038/mt.2013.154; PMID: 23831595
  • Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson DC, Fowler DH, Hardy NM, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 2013; 122:4129 - 39; http://dx.doi.org/10.1182/blood-2013-08-519413; PMID: 24055823
  • Cruz CR, Micklethwaite KP, Savoldo B, Ramos CA, Lam S, Ku S, Diouf O, Liu E, Barrett AJ, Ito S, et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 2013; 122:2965 - 73; http://dx.doi.org/10.1182/blood-2013-06-506741; PMID: 24030379
  • Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, Carrum G, Ramos C, Fayad L, Shpall EJ, et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol 2013; Forthcoming http://dx.doi.org/10.1200/JCO.2013.51.5304; PMID: 24344220
  • Dudley ME, Gross CA, Somerville RP, Hong Y, Schaub NP, Rosati SF, White DE, Nathan D, Restifo NP, Steinberg SM, et al. Randomized selection design trial evaluating CD8+-enriched versus unselected tumor-infiltrating lymphocytes for adoptive cell therapy for patients with melanoma. J Clin Oncol 2013; 31:2152 - 9; http://dx.doi.org/10.1200/JCO.2012.46.6441; PMID: 23650429
  • Besser MJ, Shapira-Frommer R, Itzhaki O, Treves AJ, Zippel DB, Levy D, Kubi A, Shoshani N, Zikich D, Ohayon Y, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res 2013; 19:4792 - 800; http://dx.doi.org/10.1158/1078-0432.CCR-13-0380; PMID: 23690483
  • Domschke C, Ge Y, Bernhardt I, Schott S, Keim S, Juenger S, Bucur M, Mayer L, Blumenstein M, Rom J, et al. Long-term survival after adoptive bone marrow T cell therapy of advanced metastasized breast cancer: follow-up analysis of a clinical pilot trial. Cancer Immunol Immunother 2013; 62:1053 - 60; http://dx.doi.org/10.1007/s00262-013-1414-x; PMID: 23595207
  • Chia WK, Teo M, Wang WW, Lee B, Ang SF, Tai WM, Chee CL, Ng J, Kan R, Lim WT, et al. Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastatic and/or locally recurrent nasopharyngeal carcinoma. Mol Ther 2014; 22:132 - 9; http://dx.doi.org/10.1038/mt.2013.242; PMID: 24297049
  • Shimizu K, Kotera Y, Aruga A, Takeshita N, Katagiri S, Ariizumi SI, Takahashi Y, Yoshitoshi K, Takasaki K, Yamamoto M. Postoperative dendritic cell vaccine plus activated T-cell transfer improves the survival of patients with invasive hepatocellular carcinoma. Hum Vaccin Immunother 2014; 10; Forthcoming http://dx.doi.org/10.4161/hv.27678; PMID: 24419174
  • Wang Z, Zhang Y, Liu Y, Wang L, Zhao L, Yang T, He C, Song Y, Gao Q. Association of myeloid-derived suppressor cells and efficacy of cytokine-induced killer cell immunotherapy in metastatic renal cell carcinoma patients. J Immunother 2014; 37:43 - 50; http://dx.doi.org/10.1097/CJI.0000000000000005; PMID: 24316555
  • Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, Vulto A, den Bakker M, Oosterwijk E, Debets R, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther 2013; 21:904 - 12; http://dx.doi.org/10.1038/mt.2013.17; PMID: 23423337
  • Davila ML, Brentjens R, Wang X, Rivière I, Sadelain M. How do CARs work?: Early insights from recent clinical studies targeting CD19. Oncoimmunology 2012; 1:1577 - 83; http://dx.doi.org/10.4161/onci.22524; PMID: 23264903
  • Kaluza KM, Vile R. Improving the outcome of adoptive cell transfer by targeting tumor escape. Oncoimmunology 2013; 2:e22059; http://dx.doi.org/10.4161/onci.22059; PMID: 23483796
  • Cao Y, Merling A, Karsten U, Schwartz-Albiez R. The fucosylated histo-blood group antigens H type 2 (blood group O, CD173) and Lewis Y (CD174) are expressed on CD34+ hematopoietic progenitors but absent on mature lymphocytes. Glycobiology 2001; 11:677 - 83; http://dx.doi.org/10.1093/glycob/11.8.677; PMID: 11479278
  • Peggs KS, Verfuerth S, Pizzey A, Khan N, Guiver M, Moss PA, Mackinnon S. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet 2003; 362:1375 - 7; http://dx.doi.org/10.1016/S0140-6736(03)14634-X; PMID: 14585640
  • Leen AM, Myers GD, Sili U, Huls MH, Weiss H, Leung KS, Carrum G, Krance RA, Chang CC, Molldrem JJ, et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med 2006; 12:1160 - 6; http://dx.doi.org/10.1038/nm1475; PMID: 16998485
  • Vanderlugt CL, Miller SD. Epitope spreading in immune-mediated diseases: implications for immunotherapy. Nat Rev Immunol 2002; 2:85 - 95; http://dx.doi.org/10.1038/nri724; PMID: 11910899
  • Vanderlugt CJ, Miller SD. Epitope spreading. Curr Opin Immunol 1996; 8:831 - 6; http://dx.doi.org/10.1016/S0952-7915(96)80012-4; PMID: 8994863
  • Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Hershkovitz L, Levy D, Kubi A, Hovav E, Chermoshniuk N, et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clin Cancer Res 2010; 16:2646 - 55; http://dx.doi.org/10.1158/1078-0432.CCR-10-0041; PMID: 20406835
  • Donia M, Junker N, Ellebaek E, Andersen MH, Straten PT, Svane IM. Characterization and comparison of “Standard” and “Young” tumor infiltrating lymphocytes for adoptive cell therapy at a Danish Translational Research Institution. Scand J Immunol 2011; Forthcoming PMID: 21955245
  • Dudley ME, Gross CA, Langhan MM, Garcia MR, Sherry RM, Yang JC, Phan GQ, Kammula US, Hughes MS, Citrin DE, et al. CD8+ enriched “young” tumor infiltrating lymphocytes can mediate regression of metastatic melanoma. Clin Cancer Res 2010; 16:6122 - 31; http://dx.doi.org/10.1158/1078-0432.CCR-10-1297; PMID: 20668005
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. OncoImmunology 2014; 3:e27297
  • 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; http://dx.doi.org/10.4161/onci.22789; PMID: 23482847
  • Besser MJ. Is there a future for adoptive cell transfer in melanoma patients?. Oncoimmunology 2013; 2:e26098; http://dx.doi.org/10.4161/onci.26098; PMID: 24353909
  • Waldron TJ, Quatromoni JG, Karakasheva TA, Singhal S, Rustgi AK. Myeloid derived suppressor cells: Targets for therapy. Oncoimmunology 2013; 2:e24117; http://dx.doi.org/10.4161/onci.24117; PMID: 23734336
  • 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; http://dx.doi.org/10.1038/onc.2011.384; PMID: 21892204
  • Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, Michaud M, Menger L, Gdoura A, Tajeddine N, et al. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene 2011; 30:1147 - 58; http://dx.doi.org/10.1038/onc.2010.500; PMID: 21151176
  • Disis ML, Schiffman K, Gooley TA, McNeel DG, Rinn K, Knutson KL. Delayed-type hypersensitivity response is a predictor of peripheral blood T-cell immunity after HER-2/neu peptide immunization. Clin Cancer Res 2000; 6:1347 - 50; PMID: 10778962
  • de Vries IJ, Bernsen MR, Lesterhuis WJ, Scharenborg NM, Strijk SP, Gerritsen MJ, Ruiter DJ, Figdor CG, Punt CJ, Adema GJ. Immunomonitoring tumor-specific T cells in delayed-type hypersensitivity skin biopsies after dendritic cell vaccination correlates with clinical outcome. J Clin Oncol 2005; 23:5779 - 87; http://dx.doi.org/10.1200/JCO.2005.06.478; PMID: 16110035
  • Wolchok JD, Chapman PB. How can we tell when cancer vaccines vaccinate?. J Clin Oncol 2003; 21:586 - 7; http://dx.doi.org/10.1200/JCO.2003.12.065; PMID: 12586792
  • Jiang J, Wu C, Lu B. Cytokine-induced killer cells promote antitumor immunity. J Transl Med 2013; 11:83; http://dx.doi.org/10.1186/1479-5876-11-83; PMID: 23536996
  • Introna M, Golay J, Rambaldi A. Cytokine Induced Killer (CIK) cells for the treatment of haematological neoplasms. Immunol Lett 2013; 155:27 - 30; http://dx.doi.org/10.1016/j.imlet.2013.09.017; PMID: 24084446
  • Sangiolo D, Mesiano G, Gammaitoni L, Leuci V, Todorovic M, Giraudo L, Cammarata C, Dell’Aglio C, D’Ambrosio L, Pisacane A, et al. Cytokine-induced killer cells eradicate bone and soft-tissue sarcomas. Cancer Res 2014; 74:119 - 29; http://dx.doi.org/10.1158/0008-5472.CAN-13-1559; PMID: 24356422
  • Le HK, Graham L, Cha E, Morales JK, Manjili MH, Bear HD. Gemcitabine directly inhibits myeloid derived suppressor cells in BALB/c mice bearing 4T1 mammary carcinoma and augments expansion of T cells from tumor-bearing mice. Int Immunopharmacol 2009; 9:900 - 9; http://dx.doi.org/10.1016/j.intimp.2009.03.015; PMID: 19336265
  • Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 2005; 11:6713 - 21; http://dx.doi.org/10.1158/1078-0432.CCR-05-0883; PMID: 16166452
  • Galluzzi L, Kepp O, Vander Heiden MG, Kroemer G. Metabolic targets for cancer therapy. Nat Rev Drug Discov 2013; 12:829 - 46; http://dx.doi.org/10.1038/nrd4145; PMID: 24113830
  • 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; http://dx.doi.org/10.4161/onci.1.2.19026; PMID: 22720239
  • 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; http://dx.doi.org/10.4161/onci.23510; PMID: 23687621
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013; 31:51 - 72; http://dx.doi.org/10.1146/annurev-immunol-032712-100008; PMID: 23157435
  • Budde LE, Berger C, Lin Y, Wang J, Lin X, Frayo SE, Brouns SA, Spencer DM, Till BG, Jensen MC, 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:e82742; http://dx.doi.org/10.1371/journal.pone.0082742; PMID: 24358223
  • Labarriere N, Fortun A, Bellec A, Khammari A, Dreno B, Saïagh S, Lang F. A full GMP process to select and amplify epitope-specific T lymphocytes for adoptive immunotherapy of metastatic melanoma. Clin Dev Immunol 2013; 2013:932318; http://dx.doi.org/10.1155/2013/932318; PMID: 24194775
  • Ma C, Cheung AF, Chodon T, Koya RC, Wu Z, Ng C, Avramis E, Cochran AJ, Witte ON, Baltimore D, et al. Multifunctional T-cell analyses to study response and progression in adoptive cell transfer immunotherapy. Cancer Discov 2013; 3:418 - 29; http://dx.doi.org/10.1158/2159-8290.CD-12-0383; PMID: 23519018
  • Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol 2012; 13:780 - 8; http://dx.doi.org/10.1038/nrm3479; PMID: 23175281
  • Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol 2010; 11:621 - 32; http://dx.doi.org/10.1038/nrm2952; PMID: 20683470
  • Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. The BCL-2 family reunion. Mol Cell 2010; 37:299 - 310; http://dx.doi.org/10.1016/j.molcel.2010.01.025; PMID: 20159550
  • Chipuk JE, Fisher JC, Dillon CP, Kriwacki RW, Kuwana T, Green DR. Mechanism of apoptosis induction by inhibition of the anti-apoptotic BCL-2 proteins. Proc Natl Acad Sci U S A 2008; 105:20327 - 32; http://dx.doi.org/10.1073/pnas.0808036105; PMID: 19074266
  • Karlsson H, Lindqvist AC, Fransson M, Paul-Wetterberg G, Nilsson B, Essand M, Nilsson K, Frisk P, Jernberg-Wiklund H, Loskog A. Combining CAR T cells and the Bcl-2 family apoptosis inhibitor ABT-737 for treating B-cell malignancy. Cancer Gene Ther 2013; 20:386 - 93; http://dx.doi.org/10.1038/cgt.2013.35; PMID: 23788110
  • Mardiros A, Dos Santos C, McDonald T, Brown CE, Wang X, Budde LE, Hoffman L, Aguilar B, Chang WC, Bretzlaff W, et al. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood 2013; 122:3138 - 48; http://dx.doi.org/10.1182/blood-2012-12-474056; PMID: 24030378
  • Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013; 19:747 - 52; http://dx.doi.org/10.1038/nm.3161; PMID: 23644516
  • Semeraro M, Galluzzi L. Novel insights into the mechanism of action of lenalidomide. OncoImmunology 2014; 3 Forthcoming
  • Noonan K, Rudraraju L, Ferguson A, Emerling A, Pasetti MF, Huff CA, Borrello I. Lenalidomide-induced immunomodulation in multiple myeloma: impact on vaccines and antitumor responses. Clin Cancer Res 2012; 18:1426 - 34; http://dx.doi.org/10.1158/1078-0432.CCR-11-1221; PMID: 22241792
  • Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Lenalidomide-based immunochemotherapy. Oncoimmunology 2013; 2:e26494; http://dx.doi.org/10.4161/onci.26494; PMID: 24482747
  • Kotla V, Goel S, Nischal S, Heuck C, Vivek K, Das B, Verma A. Mechanism of action of lenalidomide in hematological malignancies. J Hematol Oncol 2009; 2:36; http://dx.doi.org/10.1186/1756-8722-2-36; PMID: 19674465
  • Lai JP, Rosenberg AZ, Miettinen MM, Lee CC. NY-ESO-1 expression in sarcomas: A diagnostic marker and immunotherapy target. Oncoimmunology 2012; 1:1409 - 10; http://dx.doi.org/10.4161/onci.21059; PMID: 23243610
  • Suri A, Saini S, Sinha A, Agarwal S, Verma A, Parashar D, Singh S, Gupta N, Jagadish N. Cancer testis antigens: A new paradigm for cancer therapy. Oncoimmunology 2012; 1:1194 - 6; http://dx.doi.org/10.4161/onci.20686; PMID: 23170277
  • Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 2005; 5:615 - 25; http://dx.doi.org/10.1038/nrc1669; PMID: 16034368
  • O’Connell FP, Pinkus JL, Pinkus GS. CD138 (syndecan-1), a plasma cell marker immunohistochemical profile in hematopoietic and nonhematopoietic neoplasms. Am J Clin Pathol 2004; 121:254 - 63; http://dx.doi.org/10.1309/617DWB5GNFWXHW4L; PMID: 14983940
  • Chiron D, Surget S, Maïga S, Bataille R, Moreau P, Le Gouill S, Amiot M, Pellat-Deceunynck C. The peripheral CD138+ population but not the CD138- population contains myeloma clonogenic cells in plasma cell leukaemia patients. Br J Haematol 2012; 156:679 - 83; http://dx.doi.org/10.1111/j.1365-2141.2011.08904.x; PMID: 21988294
  • Kaufman HL. Vaccines for melanoma and renal cell carcinoma. Semin Oncol 2012; 39:263 - 75; http://dx.doi.org/10.1053/j.seminoncol.2012.02.011; PMID: 22595049
  • Hauschild A. Adjuvant interferon alfa for melanoma: new evidence-based treatment recommendations?. Curr Oncol 2009; 16:3 - 6; http://dx.doi.org/10.3747/co.v16i3.447; PMID: 19526078
  • Heemskerk B, Liu K, Dudley ME, Johnson LA, Kaiser A, Downey S, Zheng Z, Shelton TE, Matsuda K, Robbins PF, et al. Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2. Hum Gene Ther 2008; 19:496 - 510; http://dx.doi.org/10.1089/hum.2007.0171; PMID: 18444786
  • Kharaziha P, Ceder S, Sanchez C, Panaretakis T. Multitargeted therapies for multiple myeloma. Autophagy 2013; 9:255 - 7; http://dx.doi.org/10.4161/auto.22738; PMID: 23183549
  • Kharaziha P, De Raeve H, Fristedt C, Li Q, Gruber A, Johnsson P, Kokaraki G, Panzar M, Laane E, Osterborg A, et al. Sorafenib has potent antitumor activity against multiple myeloma in vitro, ex vivo, and in vivo in the 5T33MM mouse model. Cancer Res 2012; 72:5348 - 62; http://dx.doi.org/10.1158/0008-5472.CAN-12-0658; PMID: 22952216
  • Kharaziha P, Rodriguez P, Li Q, Rundqvist H, Björklund AC, Augsten M, Ullén A, Egevad L, Wiklund P, Nilsson S, et al. Targeting of distinct signaling cascades and cancer-associated fibroblasts define the efficacy of Sorafenib against prostate cancer cells. Cell Death Dis 2012; 3:e262; http://dx.doi.org/10.1038/cddis.2012.1; PMID: 22278289
  • Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, Schwartz B, Simantov R, Kelley S. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov 2006; 5:835 - 44; http://dx.doi.org/10.1038/nrd2130; PMID: 17016424
  • Nguyen-Hoai T, Kobelt D, Hohn O, Vu MD, Schlag PM, Dörken B, Norley S, Lipp M, Walther W, Pezzutto A, et al. HER2/neu DNA vaccination by intradermal gene delivery in a mouse tumor model: Gene gun is superior to jet injector in inducing CTL responses and protective immunity. Oncoimmunology 2012; 1:1537 - 45; http://dx.doi.org/10.4161/onci.22563; PMID: 23264900
  • Inoue M, Mimura K, Izawa S, Shiraishi K, Inoue A, Shiba S, Watanabe M, Maruyama T, Kawaguchi Y, Inoue S, et al. Expression of MHC Class I on breast cancer cells correlates inversely with HER2 expression. Oncoimmunology 2012; 1:1104 - 10; http://dx.doi.org/10.4161/onci.21056; PMID: 23170258
  • Boehrer S, Adès L, Braun T, Galluzzi L, Grosjean J, Fabre C, Le Roux G, Gardin C, Martin A, de Botton S, et al. Erlotinib exhibits antineoplastic off-target effects in AML and MDS: a preclinical study. Blood 2008; 111:2170 - 80; http://dx.doi.org/10.1182/blood-2007-07-100362; PMID: 17925489
  • de La Motte Rouge T, Galluzzi L, Olaussen KA, Zermati Y, Tasdemir E, Robert T, Ripoche H, Lazar V, Dessen P, Harper F, et al. A novel epidermal growth factor receptor inhibitor promotes apoptosis in non-small cell lung cancer cells resistant to erlotinib. Cancer Res 2007; 67:6253 - 62; http://dx.doi.org/10.1158/0008-5472.CAN-07-0538; PMID: 17616683
  • Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Fridman WH, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: DNA vaccines for cancer therapy. OncoImmunology 2014; 3:e28185 Forthcoming
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Peptide vaccines in cancer therapy. Oncoimmunology 2013; 2:e26621; http://dx.doi.org/10.4161/onci.26621; PMID: 24498550
  • Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity 2013; 39:49 - 60; http://dx.doi.org/10.1016/j.immuni.2013.07.002; PMID: 23890063
  • Hegde M, Corder A, Chow KK, Mukherjee M, Ashoori A, Kew Y, Zhang YJ, Baskin DS, Merchant FA, Brawley VS, et al. Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma. Mol Ther 2013; 21:2087 - 101; http://dx.doi.org/10.1038/mt.2013.185; PMID: 23939024
  • Kaluza KM, Kottke T, Diaz RM, Rommelfanger D, Thompson J, Vile R. Adoptive transfer of cytotoxic T lymphocytes targeting two different antigens limits antigen loss and tumor escape. Hum Gene Ther 2012; 23:1054 - 64; http://dx.doi.org/10.1089/hum.2012.030; PMID: 22734672
  • Stone JD, Kranz DM. Role of T cell receptor affinity in the efficacy and specificity of adoptive T cell therapies. Front Immunol 2013; 4:244; http://dx.doi.org/10.3389/fimmu.2013.00244; PMID: 23970885
  • Brentjens RJ. CARs and cancers: questions and answers. Blood 2012; 119:3872 - 3; http://dx.doi.org/10.1182/blood-2012-02-410373; PMID: 22538493
  • Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer 2013; 13:525 - 41; http://dx.doi.org/10.1038/nrc3565; PMID: 23880905