Review Special Focus Issue: Adoptive cell immunotherapy for cancer For reprint orders, please contact: [email protected]

Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors

Recent spectacular success in the adoptive cell therapy of leukemia and lymphoma with chimeric antigen receptor (CAR)-modified T cells raised the expectations that this therapy may be efficacious in a wide range of cancer entities. The expectations are based on the predefined specificity of CAR T cells by an antibody-derived binding domain that acts independently of the natural T-cell receptor, recognizes targets independently of presentation by the major histocompatibility complex and allows targeting toward virtually any cell surface antigen. We here discuss that targeting CAR T cells toward solid tumors faces certain circumstances critical for the therapeutic success. Targeting tumor stroma and taking advantage of TRUCK cells, in other words, CAR T cells with inducible release of a transgenic payload, are some strategies envisaged to overcome current limitations in the near future.

Hinrich Abken Clinic I Internal Medicine, Tumor Genetics, University Hospital Cologne, Cologne, Germany and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Robert-Koch-Str. 21, D-50931 Cologne, Germany hinrich.abken@ uk-koeln.de

Keywords:  adoptive cell therapy • cancer • CAR • chimeric antigen receptor • gene transfer • T cell

Concept of CAR & T cells directed for universal cytokine-mediated killing modified T cells The concept of adoptive cell therapy with chimeric antigen receptor (CAR, immunoreceptor) modified cells is based on patient’s T cells that are redirected with predefined specificity toward autologous cancer cells to execute their immune response. T cells are ex vivo modified with a recombinant receptor molecule which by the extracellular part recognizes a pre-defined target by an antibody derived binding domain and by the intra­ cellular T-cell receptor (TCR) derived signaling part initiates T-cell activation upon target engagement (Figure 1)  [1] . Such CAR T cells (also nick-named ‘T-bodies’) recognize their target independently of presentation through the major histocompatibility complex (MHC) and are thus not compromised by alterations in the antigen processing and presentation machinery as often observed during tumor progression [2,3] . CAR T cells can furthermore be modified to act as a ‘factory’ to produce a transgenic protein upon

10.2217/IMT.15.15 © 2015 Future Medicine Ltd

CAR engagement of target. Such T cell redirected for universal cytokine-mediated killing (TRUCKs) deliver in a coordinated and locally restricted fashion a ‘payload’ which may be a cytokine like IL-12 which in turn activates an innate immune response toward tumors  [4,5] . For further details we refer to recent reviews [5–7] . Compared with physiological TCR recognition, the CAR mediated T-cell activation provides several advantages in the context of adoptive cell therapy. First, CARs can redirect T cells toward a broad variety of targets as long as a binding domain with suitable specificity is available. Potential CAR targets in addition to classical MHC bound peptides also include nonclassical TCR targets such as carbohydrates, lipids and conformational epitopes. While most CARs use an antibody derived scFv for targeting, some CARs were reported, which consist of receptor ligands as binding domain fused to the trans­membrane and intracellular signaling moiety. Examples are heregulin and IL-13 mutein to target Her3/4 receptor and IL-13 receptor-α,

Immunotherapy (2015) 7(5), 535–544

part of

ISSN 1750-743X

535

Review  Abken

scFv

ζ

scFv

scFv

scFv

CD28

CD28

CD28

ζ

ζ

ζ

OX40/ 4-1BB 1o

2o

3o

CAR

TRUCK

Figure 1. Modular composition of the chimeric antigen receptor. The prototype CAR is composed of one polypeptide chain; the extracellular scFv antibody domain mediates binding to the target antigen in an MHC independent fashion. Upon CAR clustering, the intracellular TCR-derived CD3ζ chain, with or without costimulation through members of the CD28 family, initiates the downstream signaling for T-cell activation. Coreceptors may modulate CAR activity. In contrast to a first-generation (1°) CAR, second- (2°) and thirdgeneration (3°) CARs harbor one or two costimulatory moieties in their intracellular part. TRUCK cells are CAR T cells that are additionally modified with a CAR inducible expression cassette for a transgenic polypeptide product that is produced and released upon CAR signaling. Such TRUCK cells can be used as CAR targeted ‘factories’ that deposit a defined product in the targeted tissue. CAR: Chimeric antigen receptor;  scFv: Single chain fragment of variable region; TCR: T-cell receptor.

respectively [8,9] . To be recognized by CAR T cells, the antigen needs to be present on the cell surface of the targeted cell; however, TCR-like CARs were reported, which recognize intracellular antigens presented by the MHC [10,11] . In contrast to cell-bound antigen, soluble antigen, which frequently accumulates in the serum of cancer patients, does not sufficiently activate CAR T cells [12] . CAR redirected T-cell activation initiates a plethora of T-cell effector functions including cytokine release, lytic degranulation, T-cell amplification, maturation and migration. Clinical efficacy requires lasting persistence of CAR T cells which need to be protected from activation induced cell death through appropriate costimulation, for example, CD28 or 4-1BB. This is underscored by the clinical observation that patients with achieved lasting remission showed long-term CAR T-cell persistence [13] . CAR T cells can moreover provide target-specific memory which may help to prevent tumor relapse [14] . CAR signaling blocks: combined primary & costimulatory signals are superior The intracellular signaling moiety is commonly derived from the CD3ζ chain; other signaling domains including the FcɛRI γ chain are used as well. Since full T-cell activation and fine-tuning requires appropriate costimulation, the primary signaling moiety was fused to a costimulatory signaling chain, a prototype of which is CD28, giving rise to a so-called second-generation CAR [15] . Each costimulatory endodomain in the context of a CAR orchestrates a

536

Immunotherapy (2015) 7(5)

distinct pattern of effector functions as revealed by a side-by-side comparison [16] . To combine the benefits, third-generation CARs were engineered with two costimulatory domains in addition to the primary signal. This is of particular interest for redirecting T-cell subsets in more progressed maturation stages which need different costimuli for activation than young T cells [17] . Repetitive stimulation by antigen forces T cells to progress in terminal differentiation which is also the case when the CAR is engaging the cognate target. Consequently, more matured CAR T cells which lack CCR7 accumulate at the tumor lesion, at least in experimental models, while both CCR7+ and CCR7- T cells are equally efficient in targeting to the tumor lesion. Since CCR7- T cells are prone to spontaneous and activation induced cell death, which is not prevented by CD28 costimulation, the antitumor response of those cells is less efficient than that of CCR7+ T cells. Similar observations were reported for CD57+ T cells [18] . Simultaneous CD28 and OX40 costimulation reduces spontaneous and induced apoptosis of CCR7- T cells. Consequently a third-generation CD28-OX40 CAR improved the CAR-mediated antitumor activity of CCR7- T cells in the long term [17] . Since CAR T cells will progress in maturation during an antitumor attack, redirecting T cells by a CD28-OX40 CAR will improve the efficacy of adoptive cell therapy when terminal T-cell differentiation occurs. It is the particular combination of costimulatory signals to achieve the effect; the optimal combination for each T-cell subset is likely different and needs to be determined in detail.

future science group

Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors 

CAR T-cell production for clinical application Genetic equipment of T cells with CARs requires efficient gene transfer procedures and amplification of modified T cells ex vivo. While a variety of vector systems have alternatively been explored in preclinical settings, most clinical trials are currently performed using γ-retro- or lentiviral vectors. Both vectors produce stable integration of the transgene into the host genome, however, with the risk of insertional mutagenesis by provirus integration [19,20] . The retrovirus vector system has the advantage of scalable production from stable producer cells; lentivirus production uses transient systems with limited production scales, however, at high virus titers [21] . Compared to retrovirus, lentivirus vectors have a less mutagenic insertion profile and silencing is less observed. Stable integration of the transgene can also be achieved by plasmid or transposon mediated DNA transfer, both producing random integrations of plasmid DNA as concatamers [22,23] . In case of transposon-mediated DNA transfer the production procedure requires two reagents in good manufacturing practice (GMP) quality, transposase and transposon, or the in vitro selection for stable integrates in the case of plasmid transfection. In contrast to stable modifications, cells can be transiently modified with a CAR by RNA transfection that does not result in genomic integration and thereby has a high safety profile [24,25] . A clear disadvantage of RNA-modified T cells is the short time of CAR expression of some days which is even shorter after CAR ligand interaction and internalization. Consequently, multiple doses of RNA-modified T cells are needed in clinical applications [26] . The procedure to amplify CAR T cells ex vivo to clinically relevant numbers is still a matter of optimization. Most protocols are using magnetic beads coated with agonistic anti-CD3 and anti-CD28 antibodies in addition to IL-2 [20,21] . Traditionally, T cells are amplified in static culture systems which, however, require large volumes to obtain significant cell numbers. More advanced dynamic systems produce T cells in higher densities which facilitates their application in high-dose clinical trials. Most extended protocols for CAR T-cell amplification produce cells of the effector memory pheno­type. Since naive and central memory T cells persist to a greater extent than more matured T cells [27–29] , current strategies aim at shortterm amplification or enrichment of naive or central memory cells prior or after genetic engineering. Therefore common γ-chain cytokines such as IL-7, IL-15 and IL-21 are added to favor a less differentiated T-cell type  [29–31] . While these cytokines are provided in GMP quality, cell lines engineered with the costimulatory molecules represent a more physiological situa-

future science group

Review

tion (artificial APCs) [32] , however, have some GMP risks and are more difficult to handle in large-scale approaches. General problems in CAR T-cell therapy CAR immunogenicity

Most scFv’s of CARs in recent trials are of mouse origin; humoral and cellular responses against the murine domain occurred as recorded in detail in the CAR trial targeting carboanhydrase [33] . Blocking antibodies were recorded in sera of those patients; whether these antibodies substantially reduced therapeutic efficacy remains a matter of debate. The use of entirely human domains is one strategy to avoid an antibody response, however, the immunogenicity of the fusion sites of the individual CAR domains remains. The situation is even more complex since such early trials did not perform preconditioning of patients as it is generally incorporated into current trial protocols. As a consequence there is only a limited immune response shortly after adoptive cell therapy. The assumption is sustained by the observation that recent trials with preconditioning by nonmyeloablative chemotherapy did not produce a substantial humoral or cellular immune response against the CAR [34] . However, anaphylaxis resulting from RNA-modified CAR T cells was observed after third T-cell infusion, most likely through IgE antiCAR antibodies [26] , while human anti-mouse IgG antibodies, known to develop with CAR T cells, are thought to have no adverse effect. The current assumption is that anti-CAR immunogenicity may become more relevant after multiple infusions; long-term immunogenicity needs further exploration. CAR T-cell therapy-associated toxicity

While highly effective toward leukemia/lymphoma, CAR cell therapy can pose substantial risks of autoaggression against healthy tissues with cognate target antigen. While B-cell depletion after treatment with anti-CD19 CAR T cells is clinically manageable, activation of anti-ErbB2 (Her2/neu) CAR T cells against normal epithelial tissues including lung and heart resulted in patient’s death soon after adoptive transfer  [35] . The so-called ‘on-target off-organ’ activation of modified T cells by CAR engagement of target on healthy tissues is a major issue in the treatment of solid cancer and may substantially limit clinical application. In contrast to ‘off-organ’ activation, cytokine-­ associated toxicity, also known as cytokine release syndrome (CRS, ‘cytokine storm’), is not target specific and due to an extensive release of toxic levels of proinflammatory cytokines by CAR T cells which are much higher than physiologic T-cell activation would produce. The clinical reactions are common with

www.futuremedicine.com

537

Review  Abken antibody therapeutics such as nausea, fever, hypotension, vascular leakage and life-threatening multiple organ failure [36–38] . The severity of CRS seems to correlate with tumor burden, potency and dose of CAR T cells and goes along with high serum levels of IFN-γ and IL-6. The situation can be clinically reversed by application of an anti-IL6 receptor antibody, tocilizumab [38] , which is approved for several autoimmune indications, with or without co-application of nonspecific corticosteroids. Such aggressive immunosuppressive treatment, however, likely limits the therapeutic efficacy of CAR T cells. In order to minimize the risk of life threatening CRS, a system to grade the severity of CRS in individual patients and a treatment scheme based on patient characteristics, in other words, tumor burden, age, comorbidities and others, was recently set up [39] . It is currently a matter of speculation whether CRS is needed for maximum therapeutic efficacy of CAR T cells and merits further exploration. Taken together, CRS and off-tumor toxicity are severe adverse effects which could derail CAR T-cell therapy making strategies to ­eliminate CAR T cells necessary. T-cell depletion in case of toxicity

Autoreactivity seen in some patients underlines the necessity to specifically, efficiently and permanently eliminate CAR T cells from circulation. This is mostly done by expression of a ‘suicide gene’ encoding a molecule that selectively ablates genetically modified T cells. Currently, the most intensively used suicide genes are herpes simplex thymidine kinase (HSV TK) and inducible caspase 9 (iCasp9). HSV-TK has a higher affinity than the mammalian TK to specific nucleoside analogs including ganciclovir and acyclovir which upon phosphorylation incorporate into host DNA resulting in chain termination and cell death [40] . The strategy showed safe in trials and allows a gradual onset in the elimination of reactive cells [41] . A disadvantage, however, is the immunogenicity resulting in unwanted elimination of the modified T cells. The use of iCasp9 is also clinically validated and safe, and is based on dimerization of transgenic caspase 9 by a small molecule dimerizer [42] . Upon dimerization over 90% of cells are rapidly eliminated which is probably not sufficiently efficient for clinical use. Ongoing Phase I trials with CAR T cells targeting GD2 currently explore the use of iCasp9 in CAR cell therapy (NCT01822652, NCT01953900). As an alternative strategy, cells are genetically marked by a cell membrane protein which is targeted by a depleting antibody, for example, CD20 modified T cells are eliminated by anti-CD20 antibody treatment [43] . A CD34-CD20 fusion protein  [44] , which allows cell enrichment and depletion,

538

Immunotherapy (2015) 7(5)

and a truncated EGFR [45] can likewise be used. The CAR itself can be marked by a tag, for example, a myc tag, to allow elimination of modified T cells with an antitag antibody as experimentally shown for a transgenic TCR [46] . The depleting antimyc antibody, however, is so far not available for clinical use. In contrast to stable modified cells, transient CAR expression by T cells after RNA transfer will temporally limit adverse events in case of toxicity; CAR T cells disappear upon engagement of target within a few days [47] . Specific problems in targeting CAR T cells toward solid cancer While currently ongoing CAR T-cell trials for the treatment of leukemia and lymphoma offer much promise to achieve durable remission of the disease or even cure [21,37,48–50] , some trials targeting solid cancer, however, are still in an infant stage (Table 1) . The application of the CAR T-cell strategy to nonhematopoietic cancer requires the consideration of additional qualities including the disease status and tumor burden, patient preconditioning and cytokine support, CAR T-cell sensitivity to tumor mediated repression, recruitment of other arms of the patient’s immune system and stroma targeting. CAR T-cell target

The ideal target for CAR T-cell therapy is exclusively expressed on cancer cells, not on healthy cells and will produce the greatest therapeutic effect if it is critical for survival and amplification of cancer cells. These targets may be viral or mutated cell surface antigens that have mutations large enough to produce new epitopes to be recognized by an antibody. However, such target antigens are rare; most targets are also expressed by healthy cells, in some cases at lower levels, still bearing the risk of ‘on-target off-tumor’ toxicity. Such autoimmune reaction became apparent in an early phase trial targeting carboanhydrase IX to treat renal cell carcinoma, patients suffered from reversible yet discrete cholangitis and damage to bile duct epithelia due to targeting cells with physiologic carboanhydrase IX expression [51] . A fatal event occurred when a patient treated with anti-ErbB2 CAR T cells suffered from pulmonary and cardiac failure soon after T-cell administration, probably as a consequence of targeting healthy ErbB2 positive lung and heart epithelia [35] . Increasing CAR binding affinity is thought to improve targeting selectivity; experimental evidence, however, revealed that maximum T-cell activation is independent of the affinity above threshold [52] . In contrast, low-affinity CARs can more efficiently discriminate between cells with low and high antigen load than high-affinity CARs.

future science group

Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors 

Review

Table 1. Current chimeric antigen receptor T-cell trials to treat nonhematopoietic malignancies by chimeric antigen receptor modified T cells. Target

CAR

Cancer

Sponsor

Status

Clinicaltrials. gov identifier

CEA

Zeta

CEA + cancer

Cancer Research UK

Terminated

NCT01212887

CEA

CD28-zeta

CEA cancer

Roger Williams MC

Ongoing

NCT01723306

EGFR-III

 

Glioblastoma

Abramson Cancer Center

Ongoing

NCT02209376

EGFR-III

Zeta-4-1BB

EGFR + cancer

Chinese PLA General Hospital

Recruiting

NCT01869166

EGFR-III

CD28-4-1BB-zeta

Glioma

National Cancer Inst.

Recruiting

NCT01454596

ErbB2

 

ErbB2+ cancer

Chinese PLA General Hospital

Recruiting

NCT01935843

ErbB2

CD28-zeta

ErbB2+ cancer

Baylor

Recruiting

NCT00889954

ErbB2

CD28-zeta

ErbB2 glioblastoma

Baylor

Recruiting

NCT01109095

ErbB2

CD28-zeta

Sarcoma

Baylor

Recruiting

NCT00902044

ErbB2

CD28-zeta

Head and neck squamous cell cancer

King’s College London

Not yet open NCT01818323

FAP

CD28-zeta

Mesothelioma

University of Zurich

Recruiting

NCT01722149

GD2

OX40-CD28-zeta

Sarcoma, melanoma

National Cancer Inst.

Recruiting

NCT02107963

GD2

OX40-CD28-zeta

Neuroblastoma Baylor College Med.

Recruiting

NCT01822652

GD2

 

Neuroblastoma Children’s Mercy Hospital Kansas City

Recruiting

NCT01460901

GD2

OX40-CD28-zeta

Sarcoma

Baylor College Med.

Recruiting

NCT01953900

IL-13Ra2

4-1BB-zeta

Glioma

City of Hope MC

Recruiting

NCT02208362

Mesothelin

4-1BB-zeta

Mesothelin cancer

Abramson Cancer Center

Recruiting

NCT02159716

+

+

+

‘On-target off-organ’ activation may not always be harmful but also sustain the long-term therapeutic effect of CAR T cells. In the case of targeting CD19 in the treatment of leukemia, patients suffer from a lasting depletion of the B cell compartment [38,48] . Those CD19 + healthy B cells restimulate by CAR engagement the anti-CD19 CAR T cells and provide costimulatory ligands to boost the antitumor response. Since the targeted tumor and healthy cells reside in the same compartment repetitive restimulation and migration into the same niche come together to sustain activation and persistence of the anti-CD19 CAR T cells. This particular situation is not the case for the majority of solid tumors. Beside a target dependent cancer cell attack, antigen independent destruction of tumor stroma is required to control solid cancer lesions. The stroma consists of a variety of host cells including connective tissue cells, tumor fibroblasts, vascular endothelial cells and oth-

future science group

ers. Targeting connective tissue cells in solid tumors by targeting fibroblast activation protein in addition to cancer cell targeting showed superior in an experimental model [53] . A second experimental model also points to the relevance of stroma targeting for the treatment of solid cancer lesions. Destruction of large established tumors relies upon targeting of stroma cells by IFN-γ and requires strong CD28 costimulation [54] . CAR T cells thereby act by both target-dependent killing of cancer cells and by indirect mechanisms which require the action of T-cell produced IFN-γ on the tumor stroma. This is in accordance to the observation that IFN-γ is crucial in preventing tumor relapse in a syngeneic model [55] . T-cell trafficking, infiltration & repression

As a fundamental prerequisite for therapeutic efficacy, adoptively transferred T cells need to traffic to the tumor lesion. While trafficking to the sites of disease

www.futuremedicine.com

539

Review  Abken was shown in patients with leukemia [48,49,56] , only preclinical mouse data are available for T-cell targeting toward solid cancer lesions. Simultaneous bioluminescence imaging of cancer and T cells revealed the accumulation and persistence of systemically applied CAR T cells at the site of transplanted cancer [57] demonstrating that CAR T cells can traffic to solid cancer lesions. Trafficking is the result of a wide range of molecular interactions and depends on a number of soluble and cell bound factors, in particular chemokines. However, extensive amplification of modified T cells ex vivo may alter the panel of expressed chemokine receptors which are required for trafficking. Transgenic co-expression of CXCR2 (CXCL1 receptor) improves trafficking to melanoma [58] and co-expression of CCR2b the migration of CAR T cells to neuroblastoma [59] . On the other hand, the panel of chemokines produced by a variety of solid tumors does not favor T-cell infiltration into the tumor bed. In contrast, specific receptors like the endothelin B receptor prevent T-cell infiltration as shown for ovarian tumors [60] . Blocking those receptors is assumed to improve T-cell infiltration into the tumor lesion. Once cancer cell-specific T cells accumulate in the vicinity, they do not efficiently infiltrate into the tumor lesion. This became obvious when anti-ErbB2 CAR T cells did not control progression of metastases of a breast cancer patient although disseminated cancer cells were efficiently eliminated [61] . One conclusion is that CAR T cells did not sufficiently evade vasculature to penetrate the tumor tissue. This situation is aimed to be improved by targeting VEGF receptor-2, which is overexpressed by tumor-associated endothelial cells; indeed, anti-VEGF-R CAR T cells produced repression of vascularized tumors in an experimental model  [62] . Normalization of vasculature by low-dose angiogenesis inhibitors, rather than vascular destruction, also improves the antitumor efficacy [63] and may be more efficacious in the long term. The antitumor activity of CAR T cells correlates, at least to a large extend, with the persistence and sustained activation of modified T cells [37,38,48] . Conditioning chemotherapy prior adoptive cell transfer improves CAR T-cell persistence [49] . Administration of low dose IL-2 helps to improve T-cell persistence as seen in early trials using first-generation CAR T cells [64] . In addition, costimulation improves persistence of modified T cells; in particular, CD28-ζ CAR T cells persisted longer in patients than ζ CAR T cells [56] . T cells with a 4-1BB-ζ CAR persisted more than 9 months after adoptive transfer producing a significant therapeutic effect [48] . CAR T-cell persistence can also be improved by using virus specific T cells which engage viral antigens through

540

Immunotherapy (2015) 7(5)

their TCR and thereby obtain necessary costimulatory signals to survive and amplify. By means of this strategy, Epstein-Barr virus (EBV) specific, autologous T cells engineered with a CAR persisted longer in a 6-week period after infusion than T cells from the same patient and engineered with the same CAR [65] . The stage of maturation of adoptively transferred T cells also impacts their persistence in the tumor tissue. Since the frequency of CD4 + CD45RO + CD62L + memory T cells in the transferred T-cell product goes along with improved T-cell persistence [13] forthcoming trials aim at using CD62L enriched T cells for CAR modification. Once migrated into the solid tumor lesion, CAR T cells face a highly suppressive environment which is a strong barrier preventing immune surveillance [66,67] . Various cellular and soluble compartments are involved, including Treg cells, myeloid derived suppressor cells, M2-type macrophages and ligands to repressive receptors on T cells such as PD-L1, CTLA-4, B7-H family members or FasL. In this situation strategies are required to make CAR T cells more resistant to repression. For instance, the anti-tumor activity of CAR T cells in presence of Treg cells is improved in an experimental model by abrogation of IL-2 release through mutation of the CD28 CAR signaling domain [68] . Blocking the PD-1/PD-L1 may furthermore improve the antitumor activity of redirected T cells. Contact to tumor cells with PD-L1 increases PD-1 on CAR T cells [69] ; a blocking anti-PD-1 antibody improves CAR T-cell activity in a preclinical model. The recent approval of the anti-PD-1 antibody pembrolizumab by the Food and Drug Administration will move the field toward testing combination therapy of CAR T cells together with PD-1 blockade. Immune cell suppression is also mediated by cytokines like TGF-β which suppresses T-cell amplification, to a lower degree cytokine release and cytolytic activity, resulting in a reduced antitumor response in the long term. CD28 costimulation provided by a second-­generation CAR overcomes TGF-β repression resulting in a more pronounced tumor cell killing than by CD3ζ CAR T cells [70] . However, the best costimulation to tackle the complex network of immune repression still needs to be explored in relevant models and validated in clinical trials. Conclusion & future perspective So far, strategies to overcome the major hurdles for the success in the treatment of solid cancer lesions need to be empirically defined in forthcoming trials with respect to both safety and efficacy. Some modifications of the current concept are outlined below. A major goal of clinical activity in the field needs therefore to be testing CAR T cells against a broad

future science group

Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors 

range of tumors in order to optimize protocols and treatment schedules, to avoid autoimmunity and to improve persistence and function of anti-tumor T cells in the repressive tumor environment. In this context, it needs to be addressed whether one prototype CAR design can be used to drive optimal clinical responses toward the broad variety of solid tumor entities. Targeting one antigen may select for escape variants of cancer cells which give rise to relapse; simultaneous targeting of two antigens or ideally of a tumor-driver antigen will be required. Combinatorial antigen recognition is thought to improve targeting selectivity and to reduce tumor relapse and autoimmunity. T cells are engineered with two CARs recognizing two different targets on cancer cells, one CAR providing the primary signal, the other costimulation; both signals complement when both targets on cancer cells are simultaneously engaged [71] . Currently, the strategy is merely empiric and a rationale design of the co-operating CARs needs to be explored in more detail. On the other hand, outgrowing cancer cells which lost target expression makes the tumor invisible to CAR T cells. This situation is frequently seen in solid tumors; the recruitment of the innate immune system to attack those cancer cells in an antigen-independent fashion is thought to be a strategy to prevent tumor

Review

relapse after initial tumor reduction. T cells are modified with a CAR alongside with an inducible expression vector for transgenic IL-12 to release high IL-12 levels when the CAR T cells engage their cognate antigen  [4,72] . As a consequence deposited IL-12 produces a locally restricted proinflammatory response by innate cells which results in the elimination of those cancer cells which are invisible to CAR T cells [73] . Such TRUCK cells, in other words, CAR T cells engineered with an additional payload, may strengthen and fine-tune the overall immune response against solid tumors; other immune-modulating molecules need to be explored in detail to optimize the efficacy of CAR T cells toward solid tumors. Improving therapeutic efficacy of CAR T cells may require lower pretreatment intensities and lower numbers of T cells to be applied thereby potentially reducing the overall toxicities. T cells with stem celllike properties [74] may be ideal with this respect due to their strong engraftment potential after application. The general assumption is that fewer CAR T cells with a less differentiated phenotype will show similar therapeutic efficacy as high-doses of T cells with a more matured phenotype. Potentially, preconditioning may be reduced when applying those cells compared with the currently used matured central memory T cells.

Executive summary Concept of CAR-& TRUCK-modified T cells • Chimeric antigen receptor (CAR) T cells show redirected specificity toward predefined targets independently of MHC presentation; TRUCK T cells release a transgenic product in addition upon CAR engagement.

CAR signaling blocks: combined primary & costimulatory signals are superior • The costimulatory domain together with the primary T-cell receptor (TCR)-derived signal orchestrates a distinct pattern of effector functions. • The optimal combination of signals for each T-cell subset is different and needs to be determined in detail.

CAR T-cell production for clinical application • Retro- and lenti-viral vectors are commonly used to modify ex vivo patient’s T cells with CARs. • A fully automated, closed system to amplify T cells under good manufacturing practice conditions is required to establish CAR T-cell therapy for a broad panel of applications.

General problems with CAR T-cell therapy • CARs are immunogenic and provoke a humoral and cellular immune response. • CAR T-cell therapy produces some autoimmunity (on-target off-organ) which is a major challenge when targeting solid cancer. • Cytokine associated toxicity after CAR T-cell activation (cytokine storm) can be severe and life-threatening and may require immune repression and CAR T-cell elimination.

Specific problems in targeting CAR T cells toward solid cancer • A number of early phase trials are initiated to target solid cancer by CAR modified T cells. • Infiltration of solid tumor lesions and persistence of CAR T cells is insufficient and requires additional attention in the near future. • Targeting checkpoint blockade and immune repression may improve efficacy of CAR T-cell therapy in the treatment of solid cancer. • In addition to cancer cells, the tumor stroma needs to be targeted in order to reduce established large tumors.

Future perspective • Strategies to overcome the major hurdles for the success in the treatment of solid cancer lesions need to be empirically defined in forthcoming trials using a more standardized production and clinical trial protocol.

future science group

www.futuremedicine.com

541

Review  Abken Based on clinical trial data, the current situation is problematic to identify the crucial steps for therapeutic effectiveness since the design of ongoing trials is highly diverse and employs CARs of different modular composition, different T-cell populations and amplification protocols, and applies different preconditioning regimes to the patient. Head-to-head testing in multiple single parameter trials against the same target in the same disease is needed to address the issue [75] . In addition, multicenter trials with a large number of patients are required to address whether CAR T-cell therapy provides benefit with respect to the overall survival and progression-free survival compared with the standard of care. The logistic of such a trial, however, is challenging and requires the optimization of a number of issues, including the production of CAR T cells which is currently performed only in a small number of trial centers and is extremely labor and cost intensive.

Standardization and automation of these processes will help to ensure that many more centers will be able to participate in such evaluation in the near future.

References

10

Tassev DV, Cheng M, Cheung NK. Retargeting NK92 cells using an HLA-A2-restricted, EBNA3C-specific chimeric antigen receptor. Cancer Gene Ther. 19, 84–100 (2012).

11

Stewart-Jones G, Wadle A, Hombach A et al. Rational development of high-affinity T-cell receptor-like antibodies. Proc. Natl Acad. Sci. USA 106, 5784–5788 (2009).

12

Hombach A, Koch D, Sircar R et al. A chimeric receptor that selectively targets membrane-bound carcinoembryonic antigen (mCEA) in presence of soluble CEA. Gene Ther. 6, 300–304 (1999).

13

Louis CU, Savoldo B, Dotti G et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118, 6050–6056 (2011).

14

Chmielewski M, Hombach AA, Abken H. Of CARs and TRUCKs: Chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol. Rev. 257, 83–90 (2014).

Chmielewski M, Rappl G, Hombach AA et al. T cells redirected by a CD3ζ chimeric antigen receptor can establish self-antigen specific tumour protection in longterm. Gene Ther. 20, 177–186 (2013).

15

Satta A, Mezzanzanica D, Turatti F et al. Redirection of T-cell effector functions for cancer therapy: bispecific antibodies and chimeric antigen receptors. Future Oncol. 9, 527–539 (2013).

Finney HM, Lawson AD, Bebbington CR et al. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J. Immunol. 161, 2791–2797 (1998).

16

Chmielewski M, Hombach AA, Abken H. Antigenspecific T cell activation independently of the MHC: chimeric antigen receptor (CAR)-redirected T cells. Front. Immunol. 4, 71 (2013).

Hombach AA, Abken H. Costimulation by chimeric antigen receptors revisited: the T cell antitumor response benefits from combined CD28-OX40 signalling. Int. J. Cancer 129, 2935–2944 (2011).

17

Hombach AA, Chmielewski M, Rappl G et al. Adoptive immunotherapy with redirected T cells produces CCR7cells which are trapped in the periphery and benefit from combined CD28-OX40 costimulation. Hum. Gene Ther. 24, 259–269 (2013).

18

Chattopadhyay PK, Betts MR, Price DA et al. The cytolytic enzymes granyzme A, granzyme B, and perforin: expression patterns, cell distribution, and their relationship to cell maturity and bright CD57 expression. J. Leukoc. Biol. 85, 88–97 (2009).

1

2

3

4

5

6

7

8

9

542

Eshhar Z. The T-body approach: redirecting T cells with antibody specificity. Handb. Exp. Pharmacol. 181, 329–342 (2008). Elkord E, Dangoor A, Burt DJ et al. Immune evasion mechanisms in colorectal cancer liver metastasis patients vaccinated with TroVax (MVA-5T4). Cancer Immunol. Immunother. 58, 1657–1667 (2009). Garrido F, Ruiz-Cabello F, Cabrera T et al. Implications for immunosurveillance of altered HLA class I phenotypes in human tumours. Immunol. Today 18, 89–95 (1997). Chmielewski M, Kopecky C, Hombach AA et al. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 71, 5697–5706 (2011).

Muniappan A, Banapour B, Lebkowski J, Talib S. Ligandmediated cytolysis of tumor cells: use of heregulin-zeta chimeras to redirect cytotoxic T lymphocytes. Cancer Gene Ther. 7, 128–134 (2000). Kahlon KS1, Brown C, Cooper LJ et al. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res. 64, 9160–9166 (2004).

Immunotherapy (2015) 7(5)

Financial & competing interests disclosure Work in the author’s laboratory is supported by grants from the Deutsche Forschungsgemeinschaft, Bonn, Deutsche Krebshilfe, Bonn, Else Kröner-Fresenius Stiftung, Bad Homburg v.d.H., Wilhelm Sander-Stiftung, Munich, EU (European Regional Development Fund – Investing in your future), German Federal State North Rhine-Westphalia (NRW) and the Fortune Program of the Medical Faculty of the University of Cologne. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

future science group

Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors 

19

Kershaw MH et al. A Phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer Res. 12, 6106–6115 (2006).

20

Brentjens RJ et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl. Med. 5, 177ra138 (2013).

34

Till BG, Jensen MC, Wang J et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 112, 2261–2271 (2008).

35

Morgan RA, Yang JC, Kitano M et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010).

21

Porter DL, Levine BL, Kalos M et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).

36

22

Singh H et al. Manufacture of clinical-grade CD19-specific T cells stably expressing chimeric antigen receptor using Sleeping Beauty system and artificial antigen presenting cells. PLoS ONE 8, e64138 (2013).

Brentjens R, Yeh R, Bernal Y et al. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a Phase I clinical trial. Mol. Ther. 18, 666–668 (2010).

37

Kochenderfer JN, Dudley ME, Feldman SA 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 119, 2709–2720 (2012).

38

Grupp SA, Kalos M, Barrett D et al. Chimeric antigen receptor-modified T cells fo racute lymphoid leukemia. N. Engl. J. Med. 368, 1509–1518 (2013).

39

Lee DW, Gardner R, Porter DL et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124, 188–195 (2014).

40

Moolten FL. Tumor chemosensitivity conferred by inserted herpes thymidine kinase genes: paradigm for a prospective cancer control strategy. Cancer Res. 46, 5276–5281 (1986).

41

Ciceri F, Bonini C, Stanghellini MT et al. Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised Phase I-II study. Lancet Oncol. 10, 489–500 (2009).

42

Di Stasi A, Tey SK, Dotti G et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673–1683 (2011).

43

Introna M, Barbui AM, Bambacioni F et al. Genetic modification of human T cells with CD20: a strategy to purify and lyse transduced cells with anti-CD20 antibodies. Hum. Gene Ther. 11, 611–620 (2000).

44

Philip B, Kokalaki E, Mekkaoui L et al. A highly compact epitope-based marker/suicide gene for easier and safer T-cell therapy. Blood 124, 1277–1287 (2014).

45

Wang X, Chang WC, Wong CW et al. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood 118, 1255–1263 (2011).

46

Kieback E, Charo J, Sommermeyer D et al. A safeguard eliminates T cell receptor gene-modified autoreactive T cells after adoptive transfer. Proc. Natl Acad. Sci. USA 105, 623–628 (2008).

47

Birkholz K, Hombach A, Krug C et al. Transfer of mRNA encoding recombinant immunoreceptors reprograms CD4 + and CD8 + T cells for use in the adoptive immunotherapy of cancer. Gene Ther. 16, 596–604 (2009).

48

Kalos M, Levine BL, Porter DL et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 3, 95ra73 (2011).

23

Park JR et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol. Ther. 15, 825–833 (2007).

24

Zhao Y et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 70, 9053–9061 (2010).

25

Krug C, Wiesinger M, Abken H et al. A GMP-compliant protocol to expand and transfect cancer patient T cells with mRNA encoding a tumor-specific chimeric antigen receptor. Cancer Immunol. Immunother. 63, 999–1008 (2014).

26

Maus MV, Haas AR, Beatty GL et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 1, 26–31 (2013).

27

Berger C, Jensen MC, Lansdorp PM et al. Adoptive transfer of effector CD8 + T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Invest. 118, 294–305 (2008).

28

Hinrichs CS, Borman ZA, Cassard L et al. Adoptively transferred effector cells derived from naive rather than central memory CD8 + T cells mediate superior antitumor immunity. Proc. Natl Acad. Sci. USA 106, 17469–17474 (2009).

29

30

Klebanoff CA, Gattinoni L, Palmer DC et al. Determinants of successful CD8 + T-cell adoptive immunotherapy for large established tumors in mice. Clin. Cancer Res. 17, 5343–5352 (2011). Kaneko S, Mastaglio S, Bondanza A et al. IL-7 and IL-15 allow the generation of suicide gene-modified alloreactive self-renewing central memory human T lymphocytes. Blood 113, 1006–1015 (2009).

31

Pouw N, Treffers-Westerlaken E, Kraan J et al. Combination of IL-21 and IL-15 enhances tumour-specific cytotoxicity and cytokine production of TCR-transduced primary T cells. Cancer Immunol. Immunother. 59, 921–931 (2010).

32

Suhoski MM et al. Engineering artificial antigen-presenting cells to express a diverse array of co-stimulatory molecules. Mol. Ther. 15, 981–988 (2007).

33

Lamers CH, Willemsen R, van Elzakker P et al. Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. Blood 117, 72–82 (2011).

future science group

www.futuremedicine.com

Review

543

Review  Abken 49

Brentjens RJ, Rivière I, Park JH et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828 (2011).

62

Chinnasamy D, Yu Z, Theoret MR et al. Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice. J. Clin. Invest. 120, 3953–3968 (2010).

50

Kochenderfer JN, Wilson WH, Janik JE 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 116, 4099–4102 (2010).

63

51

Lamers CH, Sleijfer S, Vulto AG et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J. Clin. Oncol. 24, e20–22 (2006).

Huang Y1, Yuan J, Righi E et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl Acad. Sci. USA 109, 17561–17566 (2012).

64

Katz SC, Burga RA, McCormack E et al. Phase I HepaticImmunotherapy for Metastases study of intraarterial chimeric antigen receptor modified T cell therapy for CEA+ liver metastases.. Clin Cancer Res. pii, clincanres.1421.2014 (2015).

65

Pule MA, Savoldo B, Myers GD et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 14, 1264–1270 (2008).

66

Disis ML. Immune regulation of cancer. J. Clin. Oncol. 28, 4531–4538 (2010).

67

Whiteside TL. Immune responses to malignancies. J. Allergy Clin. Immunol. 125, 272–283 (2010).

68

Kofler DM, Chmielewski M, Rappl G et al. CD28 costimulation impairs the efficacy of a redirected T cell anti-tumor attack in the presence of regulatory T cells which can be overcome by preventing lck activation. Mol. Ther. 19, 760–767 (2011).

69

John LB, Devaud C, Duong CP et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 19, 5636–5646 (2013).

70

Koehler H, Kofler D, Hombach A et al. CD28 costimulation overcomes TGF-β mediated repression of proliferation of redirected human CD4 + and CD8 + T-cells in an anti-tumor cell attack. Cancer Res. 67, 2265–2273 (2007).

71

Kloss CC, Condomines M, Cartellieri M et al. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31, 71–75 (2013).

72

Zhang L, Kerkar SP, Yu Z et al. Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment. Mol. Ther. 19, 751–759 (2011).

73

Chmielewski M, Abken H. CAR T cells transform to trucks: chimeric antigen receptor-redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer. Cancer Immunol. Immunother. 61, 1269–1277 (2012).

74

Gattinoni L, Lugli E, Ji Y et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).

75

Hawkins RE, Gilham DE, Debets R et al. Development of adoptive cell therapy for cancer: a clinical perspective. Hum. Gene Ther. 21, 665–672 (2010).

52

53

Kakarla S, Chow KK, Mata M et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol. Ther. 21, 1611–1620 (2013).

54

Textor A, Listopad JJ, Wührmann LL et al. Efficacy of CAR T-cell therapy in large tumors relies upon stromal targeting by IFNγ. Cancer Res. 74, 6796–6805 (2014).

55

Zhang B, Karrison T, Rowley DA et al. IFN-gamma- and TNF-dependent bystander eradication of antigen-loss variants in established mouse cancers. J. Clin. Invest. 118, 1398–1404 (2008).

56

57

58

59

544

Chmielewski M, Hombach A, Heuser C et al. T cell activation by antibody-like immunoreceptors: increase in affinity of the single-chain fragment domain above threshold does not increase T cell activation against antigen-positive target cells but decreases selectivity. J. Immunol. 173, 7647–1753 (2004).

Savoldo B, Ramos CA, Liu E et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J. Clin. Invest. 121, 1822–1826 (2011). Chmielewski M, Hahn O, Rappl G et al. T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice. Gastroenterology 143, 1095–1107 (2012). Kershaw MH, Wang G, Westwood JA et al. Redirecting migration of T cells to chemokine secreted from tumors by genetic modification with CXCR2. Hum. Gene Ther. 13, 1971–1980 (2002). Craddock JA, Lu A, Bear A et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J. Immunother. 33, 780–788 (2010).

60

Kandalaft LE, Facciabene A, Buckanovich RJ et al. Endothelin B receptor, a new target in cancer immune therapy. Clin. Cancer Res. 15, 4521–4528 (2009).

61

Bernhard H, Neudorfer J, Gebhard K et al. Adoptive transfer of autologous, HER2-specific, cytotoxic T lymphocytes for the treatment of HER2-overexpressing breast cancer. Cancer Immunol. Immunother. 57, 271–280 (2008).

Immunotherapy (2015) 7(5)

future science group

Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors.

Recent spectacular success in the adoptive cell therapy of leukemia and lymphoma with chimeric antigen receptor (CAR)-modified T cells raised the expe...
3MB Sizes 0 Downloads 10 Views