Παρασκευή 28 Ιουλίου 2017

Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition,Targeting of Aberrant αvβ6 Integrin Expression in Solid Tumors Using Chimeric Antigen Receptor-Engineered T Cells,Targeting the tumor and its associated stroma,CAR T cell therapy for solid tumorsAdoptive T cell therapy of solid tumors,GSK3359609, an ICOS agonist antibody, administered alone and in combination with pembrolizumab,Dual-specific chimeric antigen receptor T cells,CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells,Mesothelin-targeted CAR-T Cells,Tumor targeted adoptively transferred T cells,T4 immunotherapy of head and neck squamous cell carcinoma using pan-ErbB targeted CAR T-cells,

Int J Biol Sci 
doi:10.7150/ijbs.14405
Review

New Strategies for the Treatment of Solid Tumors with CAR-T Cells

Hao Zhang1*, Zhen-long Ye1*, Zhen-gang Yuan1, Zheng-qiang Luo2, Hua-jun JinCorresponding address, Qi-jun qian1, 2, 3 Corresponding address
1. Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China;
2. Xinyuan Institute of Medicine and Biotechnology College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China;
3. Ningbo NO.5 Hospital (Ningbo Cancer Hospital), Ningbo 315201, China.
*These authors contributed equally to this manuscript.
This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) License. See http://ift.tt/2u49ltv for full terms and conditions.
How to cite this article:
Zhang H, Ye Zl, Yuan Zg, Luo Zq, Jin Hj, qian Qj. New Strategies for the Treatment of Solid Tumors with CAR-T Cells. Int J Biol Sci 2016; 12(6):718-729. doi:10.7150/ijbs.14405. Available from http://ift.tt/2tOT7t0

Abstract

Recent years, we have witnessed significant progresses in both basic and clinical studies regarding novel therapeutic strategies with genetically engineered T cells. Modification with chimeric antigen receptors (CARs) endows T cells with tumor specific cytotoxicity and thus induce anti-tumor immunity against malignancies. However, targeting solid tumors is more challenging than targeting B-cell malignancies with CAR-T cells because of the histopathological structure features, specific antigens shortage and strong immunosuppressive environment of solid tumors. Meanwhile, the on-target/off-tumor toxicity caused by relative expression of target on normal tissues is another issue that should be reckoned. Optimization of the design of CAR vectors, exploration of new targets, addition of safe switches and combination with other treatments bring new vitality to the CAR-T cell based immunotherapy against solid tumors. In this review, we focus on the major obstacles limiting the application of CAR-T cell therapy toward solid tumors and summarize the measures to refine this new cancer therapeutic modality.
Keywords: chimeric antigen receptor, adoptive immunotherapy, cell therapy, solid tumor.

1. Introduction

Advances in our understanding on the interaction between the immune system and tumor cells have contributed to the rapid development of novel therapeutic strategies based on chimeric antigen receptor (CAR) or T cell receptor (TCR) modified T cells. CAR-T cell therapy has achieved outstanding progresses in clinical observations, which makes it even more attractive in the development of cancer adoptive immunotherapy. The CARs endow T cells with antigen-specific recognition, activation and proliferation in a major histocompatibility complex (MHC) independent manner [1], Currently, CARs have considerably evolved to the third generation, containing two co-stimulatory molecules, such as CD28+CD134 (OX40) or CD28+CD137 (4-1BB), which have usually been demonstrated with enhanced cytokine production and tumor lytic activity and reduced activation-induced cell death (AICD) than the second or first generation CARs [2].
The emerging therapeutic approach of CAR-T cell therapy has sparked great interests, extensive studies in preclinical and clinical trials have revealed encouraging therapeutic efficacy in treating a variety of cancers, particularly in treating B-cell hematologic malignancies with CD19 CAR-T cells [3]. Nevertheless, targeting solid tumors is more challenging than targeting hematological malignancies because of tumor histopathological characteristics, shortage of specific antigens and local strong immunosuppressive microenvironment [4]. Furthermore, the on-target/off-tumor toxicity can pose significant risks. Thus, it is imperative to develop more competent and safer immunotherapy approaches by optimizing the design of CAR vectors, exploring new targets, incorporating conditional safe switches and combining with other strategies. And much work remains to be done to improve the efficacy of CAR-T cell therapy for solid tumors. It may be achieved, at least partially, by more extensive basic studies investigating the spatiotemporal dynamics of T cell activation by CARs and unraveling the connection between T cell migration in solid tumors and the effectiveness in eradication of solid tumors and metastases [5]. In this review, we discuss the current status and major obstacles for the treatment of solid tumors with CAR-T cells, thus provide some potential measures to refine this novel therapeutic modality.

2. Development and clinical application of CARs

2.1 Evolving architecture of CARs

It has been exclusively reported that CARs combine the exquisite antigen specificity of antibodies with the poly functionality and potency of cellular immunity. The unique structure of CAR endows T cells with tumor specific cytotoxicity and elevated anti-tumor activity in an MHC independent manner, through applying viral-vector technology or transposon-based system to transfect immune effector cells [6]. The classic CARs consist of an extracellular antigen recognition domain, a hinge domain, a transmembrane (TM) domain and an intracellular domain (Fig 1) [7]. The extracellular antigen-binding moiety in CARs, typically derived from a single chain variable fragment (scFv) that isolated from an antigen-specific monoclonal antibody, renders T cells the ability to bind antigens with retained specificity and affinity [8]. The hinge region mediates CAR flexibility, transduces essential signals, and exerts profound impacts on ensuring the suitable positioning of the binding domain during scFv-antigen interactions (Fig 1A) [9]. The transmembrane domains are derived from CD3-ζ, CD4, CD8, OX40, and H2-Kb [10] , and it has been clearly proved that the transmembrane domain can indeed influence the function of CAR-T cells [11]. Other investigators suggested that CAR-T cells with the CD3-ζ transmembrane domain showed more potent cytolytic activity, while CAR-T cells with CD28 transmembrane domains were more persistent [12]. A transmembrane domain from native CD3-ζ chain induces enhanced T-cell activation in comparison to mutated CD3-ζ transmembrane [13]. The intracellular domain is responsible for signal delivery within CARs, this element has been manipulated extensively in an attempt to optimize functions of engineered T cells. T cell activation relies on the phosphorylation of immune receptor tyrosine based activation motifs (ITAMs) presented in the cytoplasmic CD3-ζ domain of the TCR complex [14] (Fig 1B). The signaling domain is critical for CAR-T cells to fulfill anti-tumor functions, the construct of CARs has seen several incarnations according to the different compositions of signaling domain.
 Figure 1 Chimeric antigen receptors (CARs) architecture. (A) CARs consist of an extracellular domain, a hinge, a transmembrane domain, and an intracellular domain. The extracellular domain is typically a scFv fragment that isolated from an antigen-specific monoclonal antibody, with retained specificity and affinity. (B) The intracellular domain, derived from the phosphorylation of immunoreceptor tyrosine based activation motifs (ITAMs) presented in the cytoplasmic CD3-ζ domain of the TCR complex, transmits activation and co-stimulatory signals to T cells. (C) According to the number of signaling molecules, CARs are classified into the 1st generation (one), 2nd generation (two) and 3rd generation (three) CARs. The most applied co-stimulatory signaling molecules are CD28, 4-1BB, ICOS and OX-40.
Int J Biol Sci Image (Click on the image to enlarge.)
The first generation CARs provided the proof for the concept of the targeting and activation of CAR-T cells, but had very modest clinical activity and poor persistence in vivo [15], to overcome these limitations, the second and third generation CARs have incorporated co-stimulatory molecules, including CD27, CD28, CD134, CD137 and ICOS [16]. CAR-T cells with multiple signaling receptors have been demonstrated with sustained proliferation, enhanced cytokine production, improved tumor lytic activity, and reduced AICD both in vitro and in vivo [17] (Fig 1C). Nowadays, the second-generation CAR-T cells have been more exclusively and ubiquitously applied in clinical trials than the third generation CAR-T cells, because the reduced activation threshold of the third generation CAR-T cells may cause on-target/off-tumor side effects to normal tissues.

2.2 Therapeutic advantages of CARs technology

The use of CARs confers several advantages over TCR transgenes. Based on the MHC independent antigen recognition, CARs are able to bypass the mechanism that employed by tumors to evade immune detection through down-regulating MHC-I molecule [18]. Theoretically, CARs are able to detect almost all antigens that can be recognized by antibodies, including protein antigens, carbohydrate and lipid antigens and so on, that is, CARs are more universally applicable for immunotherapy to treat diseases [19]. In addition, the intracellular signaling domains within CARs are more flexible so that can be designed to compensate the down-regulation of co-stimulatory molecules induced by cancer cells [20]. Therefore, CAR-T cells harness maximal treatment resources for adoptive immunotherapy over TCR-T cells, for which the main hurdle is that the effect is HLA/MHC dependent.

2.3 Clinical trials utilizing CAR-T cells

The promising clinical trials have generated remarkable responses in cancer patients, which provided a solid foundation for inspiring the application of CAR-T cell based adoptive cell immunotherapy in multiple oncological settings. To date, clinical trials utilizing the second generation CD19 CAR-T cells to treat hematological malignancies have resulted in the most encouraging clinical responses [21], and the CD19-CAR T cell therapy (CTL019) has been highly appraised and approved as the breakthrough therapy by the FDA. Currently, adoptive CAR-T cells focusing on CD20 are also being evaluated in clinical trials for the treatment of B-cell malignancies, for which other CAR-T cells targeting CD22, CD30 and CD33 are now at the stage of in vitro trials [22]. At present, CAR-T cell therapy has demonstrated success as a novel treatment modality that the commercial manufacture of gene-modified T cells at industrial scale for the treatment of advanced cancers is becoming a hotspot worldwide. Mention worthy, the Juno therapeutics, Novartis and Kite are leading Big Pharmacies in the world due to their pioneering contributions to the development of CAR-T cell therapy. Figure 2 shows a flow chart of adoptive immunotherapy using CAR-T cells in clinical treatment.
In contrast to the remarkable clinical responses of CAR-T cell immunotherapy for hematologic malignancies, treating solid tumors with CAR-T cells has been limited by tumor histopathological structure and strong immunosuppressive environment, wherein the lack of ideal target is another crucial deficiency for the treatment of solid tumors. Currently the preferred therapeutic targets to treat ovarian cancer and neuroblastoma with CAR-T cells are FRα and GD2 respectively [23]. The updated statistics of therapeutic targets in solid tumor immunotherapy with CAR-T cells are showed in table 1.
 Figure 2 Schema of adoptive cellular therapy with CAR-T cells. PBLs harvested from specifically selected patients. T cells were isolated, activated and genetically modified to express a transgene encoding tumor-specific CARs. The genetically modified T cells are then expanded on a large scale using a cell processing center in vitro to a sufficient number, and thus infused back into patients, with or without chemo-radio therapeutic preconditioning.
Int J Biol Sci Image (Click on the image to enlarge.)
 Table 1 Therapeutic targets in treating solid tumors with CAR-T cells.
Target Tumor types Number of cases Clinical stage Results Citation
mesothelin
FRα
mesothelioma 4 I partial remission [24]
Lung Cancer 24 I Ongoing NCT02414269
Breast Cancer 14 I partial remission [25]
Ovarian Cancer 15 I Ongoing [26]
L1-CAM Metastatic neuroblastoma 6 I One case of partial remission; five cases of progress [27]
CAIX Metastatic renal cell carcinoma 3 I test was forced to stop because of the serious liver toxicity [28]
GD2 Neuroblastoma 11 I 5 cases of complete remission; 2 cases of partial remission; 2 cases of stable; 2 cases of tumor necrosis [29]
19 I 6 cases of complete remission; 3 cases of sick to survive; 10 cases of death [30]
FAP Malignant pleural mesothelioma 9 I Ongoing [31]
Lewis Y Bone marrow lymphoma 5 I 2 cases of stable; 1 patient died (in treatment); one case progress [32]
[33]
EGFRvIII Brain tumor 160 I/II Ongoing NCT01454596
HER2 Colon Cancer 1 I Death for off-target effects and cytokine storm syndrome [34]
HER2-positive Lung Cancer 18 I Ongoing NCT00889954
Malignant gliomas 18 I Ongoing NCT01109095
CD20 Follicular lymphoma;
Mantle cell lymphoma
7 I 2 cases of complete remission; 1 case of partial remission; 4 cases of stable [35]
3 II 2 cases of complete remission; 1 case of partial remission [36]
PSMA Prostate Cancer 18+18 I Ongoing NCT00664196NCT01140373
kLC B-cell lymphoma, CLL, multiple myeloma 18 I Ongoing NCT00881920
CD30 Hodgkin's lymphoma,NHLs 18 I Ongoing NCT01316146
CEA Stomach cancer et.al 14 I Ongoing [37]
Metastatic adenocarcinoma 48 II Ongoing NCT01723306
Metastatic Breast Cancer 26 I Ongoing NCT00673829
FRα, α-folate receptor; L1-CAM, L1-cell adhesion molecule; CAIX, carboxy-anhydrase-IX; FAP, Fibroblast activation protein; HER2, human epidermal growth factor receptor 2; CEA, carcinoembryonic antigen; PSMA, Prostate Specific Membrane Antigen; CEA, Carcino Embryonie Antigen.

3. Overcome the limiting obstacles of CAR-T cell therapy against solid tumors

CAR-T cells recognize cell surface antigens through scFv structures, which typically contain the variable domains of the light and heavy chains, in non-MHC restricted manner [38]. The membrane protein CD19 is widely expressed by almost all the B cells, and B-cell hematologic malignancies are with relatively uniform structure characteristics and so on [39]. All these properties lead to that most patients with B-cell malignancies exhibited inspiring curative effect after CD19 CAR-T cell therapy. But the application of CAR-T cell therapy in solid tumor treatment is severely limited by heterogeneity characteristics, shortage of tumor specific antigens and immunosuppressive microenvironment. Now we make an analysis on the limiting factors for the application of CAR-T cells in solid tumor treatment and discuss the relevant countermeasures.

3.1 Poor infiltration of T lymphocytes into solid tumors

Most hematologic malignancies are associated with hematopoietic stem cell regeneration dysfunction [40], without forming tissue structure. In contrast, solid tumors have special histopathological features, such as high concentration of blood vessel, wide gap of vessel wall clearance, extensive vascular leakage, poor integrity of issue structure, and so on. And these features cause selectively enhanced permeability and retention of lipid particles and macromolecular substances within solid tumors. The phenomenon of enhanced permeability and retention effect is called the EPR effect [4142]. The presence of high number of tumor-infiltrating lymphocytes (TIL) and extensive infiltration have been found as major indicators of favorable patient prognosis and positive therapeutic responses in treating several solid tumors [43], including colorectal cancer [44], lung cancer [45], and ovarian carcinomas [4647]. The EPR effect of solid tumor and the suppressive nature of the tumor microenvironment play important roles in impeding the infiltration into tumor tissues of effector T lymphocytes [4849]. Understanding and manipulating the factors contributing to the infiltration of T lymphocytes can be helpful to further improve the selective targeting of tumor tissues.
The process of T cells trafficking include rolling, adhesion, extravasation, and chemotaxis [50], and the trafficking of T cells to the tumor microenvironment is essential for the success of T cell based cancer immunotherapy. The clinical curative effect of T cell based immunotherapy against solid tumors has been more moderate than advanced melanoma or hematologic malignancies, overcoming hurdles of the migration of T cells is one of the major challenges in CAR-T cell immunotherapy, mismatching of chemokine-chemokine receptor pairs, down regulation of adhesion molecules, and aberrant vasculature may also contribute to the poor homing of T cells.
Studies have found that the CD3+, CD8+ T cell as well as B lymphocytes infiltrations are significantly correlated with the existence of tumor high endothelial venules (HEVs). Tumor HEVs are specifically located in lymphocytes concentrated areas, high density of tumor HEVs predicts low risk of relapse and metastasis [4351]. It is also an important access for lymphocyte infiltration into tumor sites, being associated with clinical prognosis makes it an indicator of tumor diagnosis and therapy. Over-expression of endothelin B receptor (ETBR) in tumor blood vessels is another limiting factor of lymphocyte infiltrating into tumor tissues through impeding the adhesion of lymphocyte to vascular endothelium [52]. And tumor angiogenesis has been found down-regulating endothelial cell-adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1) [53]. All the above limiting factors block the homing of T cells and thus impact the efficiency of tumor immunotherapy. Theoretically, ETBR blockade and ICAM-1 up-regulation could therapeutically promote T cells homing and enhance immunotherapy efficacy. ETBR inhibitor BQ-788 (a specific ETBR inhibitor peptide) has been revealed increasing T cell adhesion to human endothelium in vitro. Angiogenesis is a prerequisite for the outgrowth and metastasis of cancer cells [54], some angiogenic factors produced by tumor cells are responsible for the down-regulation of ICAM-1. For example, previous evidences have showed the up-regulation of ICAM-1 expression of human umbilical vein endothelial cells (HUVECs) after VEGF stimulation. But in contrast, prolonged stimulation (which occurs during the development of tumor) results in the down-regulation of ICAM-1 expression and leukocyte adhesion [55]. In addition, tumor necrosis factor-α (TNF-α) also induces the up-regulation of ICAM-1 [56]. Some relevant strategies to enhance the trafficking of CAR-T cells into solid tumors are discussed below.

3.1.1 Enhance CAR-T cells trafficking to tumor sites

A potential result of the special histopathological structure of solid tumor may lie in the observed lack of sufficient T cells within tumor tissues. Similarly, the insufficient migration of CAR-T cells to tumor sites also critically limits the efficacy of CAR-T cell immunotherapy against solid tumors. The limiting impact may result from unfavorable chemokine gradients, which means that tumor-specific T cells may lack the appropriate chemokine receptors for chemokines secreted by tumor cells [57].
Tumor-derived chemokines are also attractive targets for CAR-T cell immunotherapy due to their immune-modulatory effects: decrease the immunogenicity of tumors and the desensitization of chemokine receptors on T cells [58]. At the same time, tumor cells can utilize chemokines in this manner to provide autocrine growth signals and signals to enhance angiogenesis [59]. Thereby, once these tumor cells are eliminated, the remaining tumor cells will be more vulnerable. These above mechanisms envision the possibility to redirect T cells to predetermined targets through arming T cells with relevant chemokine receptors. Several studies have verified this principle through arming CAR-T cells with the expression of CXCR2 (CXCL1 receptor) [60], CCR4 (CCL17 receptor), Gro-a, CCL17 [61], and CCL2.
Nevertheless, tumor cells adopt multiple inhibitory strategies, it is challenging to derive CAR-T cells accommodating all the immune-modulatory genes which are required to overcome tumor inhibition while increasing CAR-T cell trafficking, survival, and safety [62]. Some researchers resorted to oncolytic viruses, which selectively infect, lyse, and replicate in malignant cells while sparing normal cells to solve this complex task by arming CAR-T cells with oncolytic virus expressing the chemokine RANTES and the cytokine IL-15 [63].
In addition, the difference of T cell administration also exerts an important impact on CAR-T cells expansion and effector differentiation. Studies have evaluated two different routes of CAR-T cells delivery, regional intra-pleural administration and conventional systemic intravenous of mesothelin-targeted M28z CAR-T cells, the former route presented robust T cells persistence and enhanced anti-tumor efficacy compared to the latter by circumventing obligate circulation and transient pulmonary sequestration [64]. Similarly, the intra-cerebral injection method of CAR-T cells has also been applied to treat glioblastoma with CAR-T cells to avoid the traffic blocking blood brain barrier [65]. The remarkable ability of regional delivery of CAR-T cells provides another approach to enhance functional T cell persistence and improve therapeutic efficacy through choosing favorable traffic route for CAR-T cells.

3.1.2 Cytokines released by engineered CAR-T cells promote tumor elimination

Inflammatory cells have a significant correlation with the growth and metastasis of cancer cells, indicating an improvement measure for CAR-T cell therapy by modulating tumor stroma through engineering CAR-T cells to secrete cytokines, such as interleukin-12 (IL-12) [6667]. The mechanism is that once activated by the CAR vector, T cells secrete IL-12, which can further activate innate immune cells response toward tumor cells that are invisible to CAR-T cells and subsequent inaccessible to antigen-directed immunotherapy, the process of T cells redirected for universal cytokine-mediated killing is also known as TRUCKs [68]. The TRUCKs have been demonstrated with remarkable therapeutic efficacy against tumors in pre-clinical models. So design T cells redirected by a tumor-targeting CAR and additionally engineered with a CAR-inducible cytokine cassette upon CAR engagement of cognate antigen (also termed as the fourth generation CARs by some scientists), such as the CAR-inducible IL-12 (iIL-12), which has been revealed with recruiting macrophage effect [6970]. This design can supplement the defect that CAR-T cell can't eliminate inaccessible tumor lesions, in a manner associated with reduced systemic toxicity.

3.1.3 Optimizing culture condition for CAR-T cells

Cytokine and stimulation conditions are indispensable ingredients in the process of CAR-T cells manufacturing, and several reports have indicated the influence of cytokines and growth conditions on the expansion and phenotype of immune T cells [71-73]. Thus, determining the choice of cytokines and optimizing the growth conditions are crucial for the expansion and related anti-tumor activity of CAR-T cells. The cytokines of IL-7 and IL-15 or IL-2 are mostly used as growth factors for the culture of CAR-T cells [74]. Studies have shown that IL-7 and IL-15 are superior to IL-2 for preserving CAR-T cell expansion in vitro and in vivo, CAR-T cells fed with IL-7 and IL-15 showed more sustained expansion and superior survival when exposed to serial antigens stimulation, and thus exhibited enhanced persistence and antitumor activity [7576]. In conclusion, these approaches lead to better living conditions for CAR-T cells, and can be translated into clinical immunotherapy.

3.2 Scarcity of specific antigen within solid tumors

The solid tumor heterogeneity in biological structure is a preponderant limiting factor of CAR-T cell immunotherapy for solid tumors. Tumor heterogeneity may result from subject factors and individual factors. The subject factors include the differences in cell origin and patient ethnicity, diversity that caused by genetic and epigenetic changes [77]. While the individual factors are mainly caused by tumor physiological heterogeneity among patients, intra-tumor heterogeneity, different distribution of an individual tumor, the presence of cancer stem cells or the direction of evolution [78]. Tumor heterogeneity results in that the immunotherapy target become specific to only a portion of tumor cells, which worsens the prognosis of patient and increases the recurrence and metastasis of cancer.
Therefore, the most advantageous method to treat solid tumors with CAR-T cells is to identify and project the specific cell surface antigens, but this optimal selection is severely hindered by the shortage of tumor specific antigens (TSA) under the circumstances of high heterogeneity. The posterior selection is tumor associated antigens (TAA) that relatively over expressed on the tumor cell surface, but CAR-T cells targeting TAAs may cause collateral damage to normal tissues. Therefore, new strategies improving the safety of clinical practice while maintaining the anti-tumor activity of CAR-T cells, including target tumor cell specific neoantigens that derived from somatic mutations of tumor cells (e.g. mutant EGFR variant III), target intracellular antigens (e.g.WT1, a peptide induced by Wilms' tumor gene 1), optimize CAR system with bi-signal independent pathways, apply suicide gene and other safe switches.

3.2.1 Engineered CARs targeting mutation phenotype of tumor cells

Epidermal growth factor receptor (EGFR) is a member of HER2 family, which frequently overexpressed in cancers and negatively correlated with clinical efficiency of treatment [79], and this makes it an inspiring research target. Researchers have found that 40-70% of brain tumors express mutant EGFR variant III (EGFRvIII) with a deletion of exons 2-7 of EGFR, which causes a defect in the extracellular ligand-binding domain and constitutive activation in a ligand-independent manner [8081]. Its specific expression on tumor cells, significant correlation with invasion and angiogenesis of tumors and patients' survival make EGFRvIII a novel promising target [82]. Arming polyclonal CTLs with tumor-specific TCR can avoid many obstacles in cellular immunotherapy, and this is called ''T-body'' approach [8083]. So the EGFRvIII targeting CAR system was utilized in the treatment of EGFRvIII expressing gliomas, and the generated T-body approach was able to secrete cytokines and lyse tumor cells in an EGFRvIII-dependent manner. This research brings us a new direction for CAR-T cell therapy, that is, to target specific tumor cell phenotypes induced by mutations of tumor cells.

3.2.2 Modify CARs to better target tumor associated antigens

Recognition of the peptide/MHC class I complexes endues T lymphocytes specific anti-tumor efficacy and enhanced cytokine secretion. Simultaneously, incorporation of the co-stimulatory signals or CD8+ adhesion molecules to the CARs can enhance the activation of T cells [84]. Peptide WT1, presented in the context of HLA-A* 02:01 (RMF/A2) [85], is an important immunologically validated oncogenic target with limited expression in normal tissues, while overexpressed in majority of leukemia and a wide range of solid tumors, especially mesothelioma and ovarian cancer [8687]. Therefore, CAR-T cells that targeting WT1 is a good improvement project. The modified CAR-T cells, containing the antigen recognition domain derived from fully human TCR-like ESK1 mAb (Called WT1 28z) have been found cytotoxic to primary AML bone marrow cells through targeting the intracellular oncoprotein WT1 [88]. Compared with targeting other antigens, WT1 28z CAR-T cells showed improved secretion of pro-inflammatory cytokines, such as IFN-γ, IL-2. And the therapeutic potential of TCR-like scFv CAR-T cells was proved able to be further enhanced by affinity maturation of the scFv fragment, such as TCR-like Q2L mAb derived scFv [89]. Hence, these researches indicate that improvement to the anti-tumor activity of CAR-T cells can be made through bidirectional modification of CAR-T cells to target intracellular antigens.
Mesothelin, a kind of cell surface glycoprotein with molecular weight of 40 kDa, is gaining much attention in clinical therapy of advanced solid tumors due to its high expression on the surface of numerous solid tumor cells [90]. As reported on the American Association for Cancer Research, a phase I study from Novartis and the University of Pennsylvania is ongoing, meso-CAR T cells were administered to advanced cancer patients who were no longer responding to multiple lines of prior therapies, the results indicated safety and pretty functionality in treated patients (table 1).
Mesothelin expression is also found in normal tissues, which raises a concern that meso-CAR T cells may damage healthy tissues and organs [91-93]. Researchers found some evidences showing that the meso-CAR T cells were detectable in the fluid around the heart of patients, but there were no related toxicities reported [94]. Although meso-CAR T cells program is still being evaluated, there is no doubt that mesothelin is another promising protein target and opens up the possibility that such an approach can benefit patients with various solid tumors.

3.2.3 Tuning affinity of CARs to selectively target tumor cells

In order to improve the specificity of CAR-T cells, separate dual CAR system was designed to recognize two different tumor antigens and separately transmit the first and second signals that are essential for T cell activation [95]. The co-transduced T cells maintain the therapeutic efficacy on the basis of the second generation and the third generation CARs, which empowers wider use of CAR-T cells and avoids potential safety issues.
Theoretically, the co-transduced T cells only destroy tumor cells that express both antigens (double positive) instead of these that express either antigen (single positive). However, studies have confirmed that co-transduced T cells are cytolytic against both single positive and double positive tumor cells. The most possible explanation is that the CARs transmitting the first signal have too robust stimulatory functions on T cells, and activate T cells even without the second signal. Thus in 2012, Kloss CC and colleagues reduced the affinity of the CARs that are responsible for transmitting the first signal in the dual CAR system and reported that co-transduced CAR-T cells did not exhibit cytolytic activity against single positive tumor cells but double positive tumor cells [96]. This research opened the door of adjusting the affinity to improve CAR-T cell specificity and thus safety performance.
Recently, the validity of the above strategy, through adjusting the affinity of the scFv components of CARs to selectively target tumor cells from normal cells, so as to reduce the off-target effects and improve the safety performance, was further verified. Tuning the functional affinity of CAR was performed to selectively target tumor cells overexpressing EGFR from normal cells based on the disparate density of EGFR expression [97]. The use of affinity-tuned scFv to target HER2 has found that decreasing the affinity of the scFv could significantly increase the therapeutic index of CAR-T cells [98]. Thus, another direction to improve safety performance of CAR-T cell based immunotherapy against solid tumors is tuning the functional affinity of CARs to discriminate the overexpressing tumor cells from normal tissues that express target at physiologic levels.

3.2.4 Applying suicide gene to enhance the safety of CAR-T cell therapy

Clinical application of CAR-T cells suggested that the anti-tumor efficacy is associated with some degree of toxicity [99], especially when targeting TAAs which are also expressed on normal cells. Therefore, strategies are needed to reverse any sign of toxicity effect.
Adding the “ideal” suicide gene to the construct of CARs can lead to selective ablation of gene modified T cells, thus prevent collateral damage to normal tissues or organs [100]. Therefore, CAR-T cell application with transgenic expression of one or two suicide genes is advisable, at the same time, the selective suicide gene would ensure the safety by irreversible elimination of specific part cells that are responsible for the unwanted toxicity. Currently, two validated suicide genes have been successfully used in clinical setting, herpes-simplex-thymidine-kinase (HSV-TK) and inducible-caspase-9 (iCasp9) to enhance the safety of CAR-T cell therapy against hematologic malignancies [101102]. So applying suicide gene modification to CAR-T cells may greatly increase the safety performance and clinical therapeutic efficacy.

3.3 Immunosuppressive environment within solid tumor

The clinical therapeutic efficacy of CAR-T cells in the treatment of solid tumors keeps a marginal characteristics compared with hematological malignancies, because the efficacy is significantly impeded by the strong immunosuppressive environment of solid tumors [4103]. The relative kinetics of CAR-T cells accumulation versus the rate of inactivation within solid tumors will ultimately determine the overall anti-tumor efficacy, and the balance will likely to be tumor-specific. The limiting factors that hinder T cells efficiency within solid tumor microenvironment are mainly from two aspects: 1) intrinsic microenvironment characteristics, such as hypoxia and low pH, the lack of arginine or tryptophan, inhibitory effects of tumor-derived cytokines, 2) inhibitory pathways against activated T cells, including intrinsic inhibitory pathways mediated by up-regulation of inhibitory receptors [104], intracellular inhibitory pathways to inhibit T cell receptor pathways [105], and effector functions after T cell activation [106]. Advanced generation of CAR-T cell inactivation is reversible within the solid tumor microenvironment by multiple mechanisms. Studies have showed that CAR-T cells undergo rapid loss of functional activity limited their therapeutic efficacy within solid tumor microenvironment, but this hypo-function was reversible when the CAR-T cells were isolated away from the tumor [107]. Whether this reversible characteristics can be attributed to the removal of inhibitory factors that reside in the tumor microenvironment is being investigated. Therefore, it is of great interests to optimize the efficacy of CAR-T cell therapy by combining with other treatments for solid tumors.

3.3.1 Engineer CARs to convert immunoregulatory signaling pathway

As mentioned above, solid tumors employ a variety of countermeasures to impair the function of CAR-T cells. Within the tumor microenvironment, the activated CAR-T cells are exhausted by many negative signal-regulated pathways, including cognate ligands reacting with their up-regulated inhibitory receptors expressed on T cells [108] and the lack of ligands for T cell co-stimulatory receptors [109]. Tumors exploit negative control signals to attenuate CAR-T cell responses, such as programmed death ligand 1 (PD-L1), which interacts with programmed death 1 (PD-1) expressed on activated T cells, and this exhausts CAR-T cells [110].
So developing a new engineering strategy to equip CAR-T cells with the capacity to convert tumor negative signal-regulated pathways into regulating pathways will be helpful, such as constructing a new chimera to convert cognate ligand into a ligand for T cell co-stimulation receptor by exchanging its transmembrane and cytoplasmic tail with that of CD28 or 4-1BB. The validated PD1: CD28 chimera was shown to efficiently convert PD-L1 into a co-stimulation ligand of primary human CD8+ CTL, resulting in enhanced cytokine secretion, increased proliferative capacity and augmented anti-tumor activity [111]. It is reasonable to speculate that genetic modification of CAR-T cells to express the above new type of chimera may greatly enhance the anti-tumor functions and also provide a platform to improve the clinical efficacy of CAR-T cell therapy under the immunosuppressive environment of solid tumors.

3.3.2 Engineer CARs to target stroma cells

With the exploration of solid tumor immunology, the tumor-associated stroma, occupying up to 90% of the tumor volume, has gained increasing attention for its role in initiating and sustaining tumor growth [112]. Cancer associated fibroblasts (CAFs), the principle ingredient of the tumor-associated stroma, play a preponderant influence in the formation of a highly protumorigenic and immunosuppressive microenvironment that mediates therapeutic resistance [113]. Additionally, immunotherapies with CAR-T cells targeting tumor- associated antigens (TAAs) often fail to eradicate CAFs, which support tumor progression directly through paracrine secretion of cytokines and growth factors.
Therefore, CAR-T cells targeting TAAs combine with CAR-T cells targeting CAFs may augment the anti-tumor function. The treatment by co-targeting CAFs in addition to cancer cells has been validated with significantly enhanced anti-tumor effects against solid tumors when compared with the treatment targeting CAFs or tumor cells alone. An immunotherapeutic target expressed on CAFs within a majority of solid tumors is necessary, fibroblast activation protein-α (FAP), a type 2 dipeptidyl peptidase, is a marker of a major subset of stromal cells in virtually carcinomas [114], making it an attractive therapeutic target. Studies have showed that genetically modified T cells with the expression of FAP-specific CAR can effectively recognize and kill FAP-positive tumor cells [115]. When combined with CAR-T cells targeting TAAs, FAP-specific CAR-T cells presented ever more attractive anti-tumor effects [116]. This novel combination provides another direction for solid tumor immunotherapy.

3.3.3 Combine with immune checkpoint inhibitors or cytokine expressing oncolytic virus

The field of cancer immunotherapy has considerably expanded with several new treatment options: immune checkpoint inhibitors, cancer vaccines, and adoptive T-cell immunotherapies. However, many drawbacks are still exist in efficacy, such as, immune checkpoint inhibitors are efficacious for just few patients with high mutation loaded melanoma and lung cancer, the efficacy in treating solid tumors with CAR-T cells is limited due to the unfavorable microenvironment [117118]. Thus, combination therapy of CAR-T cells with immune checkpoint inhibitors may be a solution. Recently, CAR-T cell therapy was combined with immune checkpoint inhibitors, which can create more favorable microenvironment by reducing its immunosuppressive effect to improve the efficacy of CAR-T cell therapy, and many clinical trials of combination therapy of CAR-T cells with checkpoint inhibitors are ongoing [19]. In addition to immune checkpoint inhibitors, CAR-T cell therapy has also been combined with oncolytic virus expressing the chemokine RANTES and the cytokine IL-15, and showed enhanced function of CAR-T cells by improving CAR-T cell trafficking and recruiting innate immune cells [119]. In conclusion, combination therapy of CAR-T cells with other treatments holds great potential for treating solid tumors.

4. Conclusions

CAR-T cell based immunotherapy has made a great success in treating B cell malignancies, but targeting solid tumors remains a tough task mainly due to the scarcity of TSAs and suppressive environment of solid tumors. However, researchers have been trying to improve the efficacy of CAR-T cell therapy against various solid tumors from many aspects including: 1) arming with cytokine or chemokine to enhance the infiltration of T cells and recruit other immune effectors; 2) optimizing the culture conditions to derive more potent CAR-T cells; 3) modifying the targeting system by changing the antigen recognition domain toward intracellular antigens or neo-antigens even bi-directional; 4) reducing the on-target/off tumor effect by applying dual targeting system, tuning the affinity of scFv fragments and applying safe switches; 5) combining with other treatments to eliminate tumor cells more thoroughly. Many clinical trials of CAR-T cell therapy targeting different antigens for treating solid tumors are ongoing. Nevertheless, mechanical studies on how CARs activate T cells, comparison between CAR and TCR, and optimization on each element in CARs are still needed to better apply CAR-T cell based immunotherapy in treating solid tumors.

Acknowledgements

This work was supported by the Chinese Key Project for Infectious Diseases (No. 2012ZX0002-014-005, 2013ZX10002-010-007), the Military Youth Project (No. 13QNP101), the State Project for Essential Drug Research and Development (2013ZX09102-060) and the Chinese State 863 projects (No. 2012AA020806).

Author contribution

Zhang H and Ye Z wrote the manuscript. Zhang H, Ye Z, Jin H and Qian Q conceived the study and revised the manuscript. Gang Y and Luo Z assisted searching the literature and provided conceptual input. All the authors participated in the discussion of the study, have read and approved the final manuscript.

Competing Interests

The authors have declared that no competing interest exists.

References

1. Bouhassira DC, Thompson JJ, Davila ML. Using gene therapy to manipulate the immune system in the fight against B-cell leukemias. Expert opinion on biological therapy. 2015;15:403-16
2. Han EQ, Li X, Wang C, Li T, Han S. Chimeric antigen receptor-engineered T cells for cancer immunotherapy: progress and challenges. J Hematol Oncol.2013;6:47
3. Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014;123:2625-35
4. Beavis PA, Slaney CY, Kershaw MH, Gyorki D, Neeson PJ, Darcy PK. Reprogramming the tumor microenvironment to enhance adoptive cellular therapy. Seminars in immunology: Elsevier. 2015
5. Chmielewski M, Hombach A, Heuser C, Adams GP, Abken H. 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. The Journal of Immunology. 2004;173:7647-53
6. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. cell. 2011;144:646-74
7. Whilding LM, Maher J. CAR T-cell immunotherapy: The path from the by-road to the freeway?. Molecular oncology. 2015;9:1994-2018
8. Shi H, Sun M, Liu L, Wang Z. Chimeric antigen receptor for adoptive immunotherapy of cancer: latest research and future prospects. Molecular cancer.2014;13:219
9. Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, Rader C. et alThe non-signaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer immunology research. 2014 canimm.0127.2014
10. Harris DT, Kranz DM. Adoptive T Cell Therapies: A Comparison of T Cell Receptors and Chimeric Antigen Receptors. Trends in Pharmacological Sciences. 2015
11. Dotti G, Gottschalk S, Savoldo B, Brenner MK. Design and development of therapies using chimeric antigen receptor-expressing T cells. Immunological reviews. 2014;257:107-26
12. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G. et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. The Journal of clinical investigation. 2011;121:1822
13. Bridgeman JS, Hawkins RE, Bagley S, Blaylock M, Holland M, Gilham DE. The optimal antigen response of chimeric antigen receptors harboring the CD3ζ transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex. The Journal of Immunology. 2010;184:6938-49
14. Kershaw MH, Westwood JA, Slaney CY, Darcy PK. Clinical application of genetically modified T cells in cancer therapy. Clinical & Translational Immunology.2014;3:e16
15. Kenderian SS, Ruella M, Gill S, Kalos M. Chimeric antigen receptor T-cell therapy to target hematologic malignancies. Cancer research. 2014;74:6383-9
16. Zhong X-S, Matsushita M, Plotkin J, Riviere I, Sadelain M. Chimeric Antigen Receptors Combining 4-1BB and CD28 Signaling Domains Augment PI3kinase/AKT/Bcl-XL Activation and CD8+ T Cell-mediated Tumor Eradication. Molecular Therapy. 2010;18:413-20
17. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M. et al4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nature medicine. 2015
18. Catalán E, Charni S, Jaime P, Aguiló JI, Enríquez JA, Naval J. et al. MHC-I modulation due to changes in tumor cell metabolism regulates tumor sensitivity to CTL and NK cells. Oncoimmunology. 2015;4:e985924
19. Hillerdal V, Essand M. Chimeric Antigen Receptor-Engineered T Cells for the Treatment of Metastatic Prostate Cancer. BioDrugs. 2015;29:75-89
20. Duong CP, Yong CS, Kershaw MH, Slaney CY, Darcy PK. Cancer immunotherapy utilizing gene-modified T cells: from the bench to the clinic. Molecular immunology. 2015
21. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. New England Journal of Medicine. 2014;371:1507-17
22. Boulassel M-R, Galal A. Immunotherapy for B-Cell Neoplasms using T Cells expressing Chimeric Antigen Receptors: From antigen choice to clinical implementation. Sultan Qaboos University medical journal. 2012;12:273
23. Singh N, Liu X, Hulitt J, Jiang S, June CH, Grupp SA. et al. Nature of tumor control by permanently and transiently modified GD2 chimeric antigen receptor T cells in xenograft models of neuroblastoma. Cancer Immunol Res. 2014;2:1059-70
24. Maus MV, Haas AR, Beatty GL, Albelda SM, Levine BL, Liu X. et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer immunology research. 2013;1:26-31
25. Hasegawa K, Nakamura T, Harvey M, Ikeda Y, Oberg A, Figini M. et al. The use of a tropism-modified measles virus in folate receptor-targeted virotherapy of ovarian cancer. Clinical Cancer Research. 2006;12:6170-8
26. Kandalaft LE, Powell Jr DJ, Coukos G. A phase I clinical trial of adoptive transfer of folate receptor-alpha redirected autologous T cells for recurrent ovarian cancer. J Transl Med. 2012;10:157
27. Park JR, DiGiusto DL, Slovak M, Wright C, Naranjo A, Wagner J. et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Molecular Therapy. 2007;15:825-33
28. Lamers CH, Langeveld SC, Groot-van Ruijven CM, Debets R, Sleijfer S, Gratama JW. Gene-modified T cells for adoptive immunotherapy of renal cell cancer maintain transgene-specific immune functions in vivo. Cancer Immunology, Immunotherapy. 2007;56:1875-83
29. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G. et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nature medicine. 2008;14:1264-70
30. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD. et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050-6
31. Petrausch U, Schuberth PC, Hagedorn C, Soltermann A, Tomaszek S, Stahel R. et al. Re-directed T cells for the treatment of fibroblast activation protein (FAP)-positive malignant pleural mesothelioma (FAPME-1). BMC cancer. 2012;12:615
32. Peinert S, Prince H, Guru P, Kershaw M, Smyth M, Trapani J. et al. Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther. 2010;17:678-86
33. Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K. et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Molecular Therapy. 2013;21:2122-9
34. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Molecular Therapy. 2010;18:843-51
35. Zhao Y, Moon E, Carpenito C, Paulos CM, Liu X, Brennan AL. et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer research. 2010;70:9053-61
36. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG. et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood. 2012;119:3940-50
37. Guest RD, Kirillova N, Mowbray S, Gornall H, Rothwell DG, Cheadle EJ. et al. Definition and application of good manufacturing process-compliant production of CEA-specific chimeric antigen receptor expressing T-cells for phase I/II clinical trial. Cancer immunology, immunotherapy: CII. 2014;63:133-45
38. Sadelain M. T-cell engineering for cancer immunotherapy. The Cancer Journal. 2009;15:451-5
39. Gill S, Porter DL. CAR-modified anti-CD19 T cells for the treatment of B-cell malignancies: rules of the road. Expert opinion on biological therapy. 2014;14:37-49
40. Trifilio S, Bennett C, Yarnold P, McKoy J, Parada J, Mehta J. et al. Breakthrough zygomycosis after voriconazole administration among patients with hematologic malignancies who receive hematopoietic stem-cell transplants or intensive chemotherapy. Bone marrow transplantation. 2007;39:425-9
41. Fang J, Nakamura H, Maeda H. The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced drug delivery reviews. 2011;63:136-51
42. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. Journal of controlled release. 2000;65:271-84
43. Martinet L, Garrido I, Filleron T, Le Guellec S, Bellard E, Fournie J-J. et al. Human solid tumors contain high endothelial venules: association with T-and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer research. 2011;71:5678-87
44. Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R. et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. New England Journal of Medicine. 2005;353:2654-66
45. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62-8
46. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer.New England Journal of Medicine. 2003;348:203-13
47. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K. et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proceedings of the National Academy of Sciences. 2007;104:3360-5
48. Yin H, Liao L, Fang J. Enhanced permeability and retention (EPR) effect based tumor targeting: the concept, application and prospect. JSM Clin Oncol Res.2014;2:1010-4
49. Rahir G, Moser M. Tumor microenvironment and lymphocyte infiltration. Cancer Immunology, Immunotherapy. 2012;61:751-9
50. Slaney CY, Kershaw MH, Darcy PK. Trafficking of T Cells into Tumors. Cancer research. 2014;74:7168-74
51. Martinet L, Le Guellec S, Filleron T, Lamant L, Meyer N, Rochaix P. et al. High endothelial venules (HEVs) in human melanoma lesions: major gateways for tumor-infiltrating lymphocytes. Oncoimmunology. 2012;1:829-39
52. Buckanovich RJ, Facciabene A, Kim S, Benencia F, Sasaroli D, Balint K. et al. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nature medicine. 2008;14:28-36
53. Dirkx AE, oude Egbrink MG, Kuijpers MJ, van der Niet ST, Heijnen VV, Bouma-ter Steege JC. et al. Tumor angiogenesis modulates leukocyte-vessel wall interactions in vivo by reducing endothelial adhesion molecule expression. Cancer research. 2003;63:2322-9
54. Griffioen AW, Molema G. Angiogenesis: potentials for pharmacologic intervention in the treatment of cancer, cardiovascular diseases, and chronic inflammation. Pharmacological reviews. 2000;52:237-68
55. Griffioen AW, Damen CA, Martinotti S, Blijham GH, Groenewegen G. Endothelial intercellular adhesion molecule-1 expression is suppressed in human malignancies: the role of angiogenic factors. Cancer research. 1996;56:1111-7
56. J Tuorkey M, K Abdul-Aziz K, A Zidan A-A. Active immunization against tumor necrosis factor-alpha decreases proinflammatory cytokines, oxidative stress mediators and adhesion molecules risk factors in streptozotocin-induced diabetic rats. Endocrine, Metabolic & Immune Disorders-Drug Targets (Formerly Current Drug Targets-Immune, Endocrine & Metabolic Disorders). 2013;13:269-74
57. Karin N, Wildbaum G. The Role of Chemokines in Shaping the Balance Between CD4+ T Cell Subsets and Its Therapeutic Implications in Autoimmune and Cancer Diseases. Frontiers in immunology. 2015:6
58. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. cell. 1994;76:301-14
59. Kershaw MH, Wang G, Westwood JA, Pachynski RK, Tiffany HL, Marincola FM. et al. Redirecting migration of T cells to chemokine secreted from tumors by genetic modification with CXCR2. Hum Gene Ther. 2002;13:1971-80
60. Craddock JA, Lu A, Bear A, Pule M, Brenner MK, Rooney CM. et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. Journal of immunotherapy (Hagerstown, Md: 1997). 2010;33:780
61. Di Stasi A, De Angelis B, Rooney CM, Zhang L, Mahendravada A, Foster AE. et al. T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting CD30 have improved homing and antitumor activity in a Hodgkin tumor model. Blood. 2009;113:6392-402
62. Curran KJ, Pegram HJ, Brentjens RJ. Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. The journal of gene medicine. 2012;14:405-15
63. Nishio N, Diaconu I, Liu H, Cerullo V, Caruana I, Hoyos V. et al. Armed Oncolytic Virus Enhances Immune Functions of Chimeric Antigen Receptor-Modified T Cells in Solid Tumors. Cancer research. 2014;74:5195-205
64. Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J. et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Science translational medicine. 2014;6:261ra151-261ra151
65. Choi BD, Suryadevara CM, Gedeon PC, Herndon II JE, Sanchez-Perez L, Bigner DD. et al. Intracerebral delivery of a third generation EGFRvIII-specific chimeric antigen receptor is efficacious against human glioma. Journal of Clinical Neuroscience. 2014;21:189-90
66. Pegram HJ, Lee JC, Hayman EG, Imperato GH, Tedder TF, Sadelain M. et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012;119:4133-41
67. Kerkar SP, Muranski P, Kaiser A, Boni A, Sanchez-Perez L, Yu Z. et al. Tumor-specific CD8+ T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts. Cancer research. 2010;70:6725-34
68. 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. Immunological reviews. 2014;257:83-90
69. Chmielewski M, Kopecky C, Hombach AA, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer research. 2011;71:5697-706
70. Zhang L, Kerkar SP, Yu Z, Zheng Z, Yang S, Restifo NP. et al. Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment. Molecular Therapy. 2011;19:751-9
71. Gargett T, Brown MP. Different cytokine and stimulation conditions influence the expansion and immune phenotype of third-generation chimeric antigen receptor T cells specific for tumor antigen GD2. Cytotherapy. 2015;17:487-95
72. Porter DL, Levine BL, Bunin N, Stadtmauer EA, Luger SM, Goldstein S. et al. A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood. 2006;107:1325-31
73. Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R. et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. New England Journal of Medicine. 2013;369:134-44
74. Ma A, Koka R, Burkett P. Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis. Annu Rev Immunol. 2006;24:657-79
75. Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR. CD19-T cells and are preserved by IL-7 and IL-15. Blood. 2014;123:3750-9
76. Caserta S, Alessi P, Basso V, Mondino A. IL-7 is superior to IL-2 for ex vivo expansion of tumour-specific CD4+ T cells. European journal of immunology.2010;40:470-9
77. Shen H, Laird PW. Interplay between the cancer genome and epigenome. cell. 2013;153:38-55
78. Roessler S, Budhu A, Wang XW. Deciphering cancer heterogeneity: the biological space. Frontiers in cell and developmental biology. 2014:2
79. Panousis C, Rayzman V, Johns T, Renner C, Liu Z, Cartwright G. et al. Engineering and characterisation of chimeric monoclonal antibody 806 (ch806) for targeted immunotherapy of tumours expressing de2-7 EGFR or amplified EGFR. British journal of cancer. 2005;92:1069-77
80. Ohno M, Natsume A, Ichiro Iwami K, Iwamizu H, Noritake K, Ito D. et al. Retrovirally engineered T-cell-based immunotherapy targeting type III variant epidermal growth factor receptor, a glioma-associated antigen. Cancer science. 2010;101:2518-24
81. Sugawa N, Ekstrand AJ, James CD, Collins VP. Identical splicing of aberrant epidermal growth factor receptor transcripts from amplified rearranged genes in human glioblastomas. Proceedings of the National Academy of Sciences. 1990;87:8602-6
82. Yamazaki H, Ohba Y, Tamaoki N, Shibuya M. A deletion mutation within the ligand binding domain is responsible for activation of epidermal growth factor receptor gene in human brain tumors. Cancer science. 1990;81:773-9
83. Eshhar Z. The T-body approach: redirecting T cells with antibody specificity. Therapeutic Antibodies. Springer. 2008 p. 329-42
84. Watanabe K, Terakura S, Martens AC, van Meerten T, Uchiyama S, Imai M. et al. Target Antigen Density Governs the Efficacy of Anti-CD20-CD28-CD3 ζ Chimeric Antigen Receptor-Modified Effector CD8+ T Cells. The Journal of Immunology. 2015;194:911-20
85. Veomett N, Dao T, Liu H, Xiang J, Pankov D, Dubrovsky L. et al. Therapeutic efficacy of an Fc-enhanced TCR-like antibody to the intracellular WT1 oncoprotein. Clinical Cancer Research. 2014;20:4036-46
86. Krug LM, Dao T, Brown AB, Maslak P, Travis W, Bekele S. et al. WT1 peptide vaccinations induce CD4 and CD8 T cell immune responses in patients with mesothelioma and non-small cell lung cancer. Cancer Immunology, Immunotherapy. 2010;59:1467-79
87. Dohi S, Ohno S, Ohno Y, Takakura M, Kyo S, Soma G-I. et al. WT1 peptide vaccine stabilized intractable ovarian cancer patient for one year: a case report.Anticancer research. 2011;31:2441-5
88. Rafiq S, Dao T, Liu C, Scheinberg DA, Brentjens RJ. Engineered T cell receptor-mimic antibody,(TCRm) chimeric antigen receptor (CAR) T cells against the intracellular protein Wilms tumor-1 (WT1) for treatment of hematologic and solid cancers. Blood. 2014;124:2155 -
89. Zhao Q, Ahmed M, Tassev D, Hasan A, Kuo T, Guo H. et alAffinity maturation of T-cell receptor-like antibodies for Wilms tumor 1 peptide greatly enhances therapeutic potential. Leukemia. 2015
90. Tchou J, Wang L-C, Selven B, Zhang H, Conejo-Garcia J, Borghaei H. et al. Mesothelin, a novel immunotherapy target for triple negative breast cancer. Breast cancer research and treatment. 2012;133:799-804
91. Servais EL, Colovos C, Rodriguez L, Bograd AJ, Nitadori J-i, Sima C. et al. Mesothelin overexpression promotes mesothelioma cell invasion and MMP-9 secretion in an orthotopic mouse model and in epithelioid pleural mesothelioma patients. Clinical Cancer Research. 2012;18:2478-89
92. Rizk NP, Servais EL, Tang LH, Sima CS, Gerdes H, Fleisher M. et al. Tissue and serum mesothelin are potential markers of neoplastic progression in Barrett's associated esophageal adenocarcinoma. Cancer Epidemiology Biomarkers & Prevention. 2012;21:482-6
93. Kachala SS, Bograd AJ, Villena-Vargas J, Suzuki K, Servais EL, Kadota K. et al. Mesothelin overexpression is a marker of tumor aggressiveness and is associated with reduced recurrence-free and overall survival in early-stage lung adenocarcinoma. Clinical Cancer Research. 2014;20:1020-8
94. Pastan I, Hassan R. Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer research. 2014;74:2907-12
95. Wilkie S, van Schalkwyk MC, Hobbs S, Davies DM, van der Stegen SJ, Pereira ACP. et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. Journal of clinical immunology. 2012;32:1059-70
96. Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nature biotechnology. 2013;31:71-5
97. Caruso HG, Hurton LV, Najjar A, Rushworth D, Ang S, Olivares S. et al. Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity. Cancer research. 2015;75:3505-18
98. Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC. et al. Affinity-Tuned ErbB2 or EGFR Chimeric Antigen Receptor T Cells Exhibit an Increased Therapeutic Index against Tumors in Mice. Cancer research. 2015;75:3596-607
99. Casucci M, Bondanza A. Suicide gene therapy to increase the safety of chimeric antigen receptor-redirected T lymphocytes. Journal of Cancer. 2011;2:378
100. Jones BS, Lamb LS, Goldman F, Di Stasi A. Improving the safety of cell therapy products by suicide gene transfer. Frontiers in pharmacology. 2014:5
101. Gargett T, Brown MP. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells.Frontiers in pharmacology. 2014:5
102. Ciceri F, Bonini C, Stanghellini MTL, Bondanza A, Traversari C, Salomoni M. 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. The lancet oncology. 2009;10:489-500
103. Shiao SL, Ganesan AP, Rugo HS, Coussens LM. Immune microenvironments in solid tumors: new targets for therapy. Genes & development. 2011;25:2559-72
104. Park HJ, Kusnadi A, Lee E-J, Kim WW, Cho BC, Lee IJ. et al. Tumor-infiltrating regulatory T cells delineated by upregulation of PD-1 and inhibitory receptors. Cellular immunology. 2012;278:76-83
105. Zheng Y, Zha Y, Gajewski TF. Molecular regulation of T-cell anergy. EMBO reports. 2008;9:50-5
106. Moon EK, Wang L-C, Dolfi DV, Wilson CB, Ranganathan R, Sun J. et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clinical Cancer Research. 2014;20:4262-73
107. Beatty GL, Moon EK. Chimeric antigen receptor T cells are vulnerable to immunosuppressive mechanisms present within the tumor microenvironment.Oncoimmunology. 2014;3:e970027
108. Gajewski TF, Meng Y, Blank C, Brown I, Kacha A, Kline J. et al. Immune resistance orchestrated by the tumor microenvironment. Immunological reviews.2006;213:131-45
109. Anderson RC, Anderson DE, Elder JB, Brown MD, Mandigo CE, Parsa AT. et al. Lack of B7 expression, not human leukocyte antigen expression, facilitates immune evasion by human malignant gliomas. Neurosurgery. 2007;60:1129-36
110. Tewalt EF, Cohen JN, Rouhani SJ, Guidi CJ, Qiao H, Fahl SP. et al. Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells. Blood. 2012;120:4772-82
111. Prosser ME, Brown CE, Shami AF, Forman SJ, Jensen MC. Tumor PD-L1 co-stimulates primary human CD8+ cytotoxic T cells modified to express a PD1: CD28 chimeric receptor. Molecular immunology. 2012;51:263-72
112. Santos AM, Jung J, Aziz N, Kissil JL, Puré E. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. The Journal of clinical investigation. 2009;119:3613
113. Kakarla S, Chow KK, Mata M, Shaffer DR, Song X-T, Wu M-F. et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Molecular Therapy. 2013;21:1611-20
114. Saadi A, Shannon NB, Lao-Sirieix P, O'Donovan M, Walker E, Clemons NJ. et al. Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers. Proceedings of the National Academy of Sciences. 2010;107:2177-82
115. Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO. et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-α. Science. 2010;330:827-30
116. Lo A, Wang L-CS, Scholler J, Monslow J, Avery D, Newick K. et alTumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer research. 2015 canres.3041.2014
117. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62-8
118. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69-74
119. Nishio N, Dotti G. Oncolytic virus expressing RANTES and IL-15 enhances function of CAR-modified T cells in solid tumors. Oncoimmunology.2015;4:e988098

Author contact

Corresponding address Corresponding authors: Qi-Jun Qian, Tel: + 86-21-81875371; Fax: + 86-21-65580677; E-mail: qianqj@sino-gene.cn; Hua-Jun Jin, Tel: + 86-21-81875372; E-mail: hj-jin@hotmail.com.

Logo of nihpa About Author manuscripts Submit a manuscript HHS Public Access; Author Manuscript; Accepted for publication in peer reviewed journal;
PMCID: PMC4050065
NIHMSID: NIHMS592410

CAR T cells for solid tumors: armed and ready to go?

Sunitha Kakarla, PhD, Scientist and Stephen Gottschalk, MD, Professor
Sunitha Kakarla, 
Corresponding author: Stephen Gottschalk Center for Cell and Gene Therapy Baylor College of Medicine 1102 Bates Street, Suite 1770 Houston, TX 77030 Phone: 832-824-4179 Fax: 832-825-4732 gro.hcxt@csttogms

Introduction

T cells - the armed forces of our immune repertoire are constantly coursing through the bloodstream on the lookout for foreign antigens. On encountering a solid tumor - a complex and dynamic mass composed of rogue host cells, T cells often fail to mount an effective response. Even when tumor responsive T cells extravasate to tumor sites, they are faced with a barrage of immunosuppressive factors that render them non-responsive. Thus, even the most adroit natural eliminator is exhausted in the tumor environment. Fortunately, there exist genetic approaches that can reprogram ordinary circulating T cells to highly specific slayers of cancer cells.-
Genetically modifying T cells with chimeric antigen receptors (CARs) is the most commonly used approach to generate tumor specific T cells. As outlined in detail in other contributions to this themed journal issue, CARs consist of an ectodomain, commonly derived from a single chain variable fragment (scFv), a hinge, a transmembrane domain, and an endodomain with one (1st generation), two (2ndgeneration), or three (3rd generation) signaling domains derived from CD3Ζ and/or co-stimulatory molecules. While CAR T cells have been successful in early phase clinical studies treating CD19-positive hematological malignancies,- the success of CARs in the purview of solid tumors has been greatly hampered by the lack of unique tumor associated antigens, inefficient homing of T cells to tumor sites and the immunosuppressive microenvironment of solid tumors. In this review we delineate the barriers imposed by solid tumors on CARs and strategies that have and should be undertaken to improve therapeutic response.

Solid tumor antigens

Tumor associated antigens can be divided into several groups including antigens that 1) contain novel peptide sequences due to gene mutation, 2) are expressed in a tissue/lineage specific fashion, 3) are normally expressed during fetal development or at immunoprivileged sites, or 4) are expressed at higher than normal levels on tumor cells compared to non-malignant host cells.

Mutated antigens

Ideally, the targeted antigen should contain a novel peptide sequence, limiting its expression to tumor cells. One example is a splice variant of the epidermal growth factor receptor (EGFRvIII). Other antigens that may provide some target exclusivity include those with altered post-translational modifications or those that present conformational epitopes unique to the tumor microenvironment. Examples for the former include abnormal, tumor-specific glycosylation patterns of MUC-1, and for the latter include i) conformational EGFR and erythropoietin-producing hepatocellular A2 receptor (EphA2) epitopes.;

Tissue/Lineage antigens

Tissue/Lineage restricted antigens by definition show restrictive expression patterns and may seem particularly attractive. While tissue expendability is acceptable or correctable in certain conditions like B-cell aplasia with CD19 CART cells, the function of most solid organs cannot be readily replaced. However, tissue-specific antigens of organs that are dispensable such as prostate specific cancer antigen (PSCA), are actively being explored.

Developmental antigens

Antigens expressed during fetal development or at immunoprivileged sites such MAGE family members or NY-ESO-1 are actively being targeted with αβ TCRs.; However most of these antigens are in cytoplasm, making them inaccessible to scFv-based CARs that recognize antigens expressed on the cell surface. However, scFv have been developed that recognize peptide derived from cytoplasmic proteins in the context of HLA molecule, making them ‘CAR targetable’.

Overexpressed antigens

The majority of targeted antigens are only overexpressed in comparison to normal tissues, raising concerns about ‘on target/off tumor’ side effects, which in most cases cannot be adequately assessed in murine models. Nevertheless, a broad array of overexressed solid tumor antigens has been targeted in preclinical models with CAR T cells (Table 1) and several clinical studies have been conducted (Table 2).
Table 1
Solid tumor antigens for CAR T-cell therapy
Table 2
Clinical studies with CAR T cells targeting solid antigens

Targeting multiple antigens

In contrast to conventional T cells that only recognize single antigens, CAR T cells can be genetically modified to recognize multiple antigens, which should allow the recognition of unique antigen expression patterns on tumor cells. One example include ‘split signal CARs’ that limit full T-cell activation to tumors expressing multiple antigens.- Other strategies to target multiple antigens include tandem CARs (TanCARs), which contain ectodomains with two scFvs, and so called ‘universal ectodomain CARs’ that incorporate avidin or a FITC-specific scFv to recognize tumor cells that have been incubated with tagged MAbs.; Targeting multiple antigens should also limit the risk of immune escape.
In summary, numerous solid tumor antigens are being actively explored for CAR T-cell therapy, however only few are uniquely tumor specific. While genetic approaches can be used to increase specificity, there is a growing exigency to discover novel tumor antigens as CAR T cells become more potent.

Clinical trials with solid tumor-specific CAR T cells

Pre-clinical studies have targeted a broad array of solid tumor antigens with CAR T cells (Table 1). Antigens currently targeted in clinical studies include carbonic anhydrase IX (CAIX), CD171, folate receptor alpha (FR-α), GD2, human epidermal growth factor receptor 2 (HER2), mesothelin, EGFRvIII, fibroblast activation protein (FAP), carcinoembryonic antigen (CEA), and vascular endothelial growth factor receptor 2 (VEGF-R2) (Table 2). The majority of clinical studies so far have used 1st generation CAR T cells, and while the studies have demonstrated feasibility, the clinical results have been in general disappointing.- Nevertheless, these studies gave important insights into CAR T-cell biology. Lamers et al. infused renal carcinoma patients with polyclonal T cells expressing a 1stgeneration CAIX-specific CAR, and observed ‘on target/off cancer’ side effects, and the development of anti-CAR immune responses resulting in limited T-cell persistence. Subsequently, pretreatment with CAIX MAbs prior to CAR T-cell transfer prevented hepatitis and abrogated the induction of anti-CAR immune responses.
Limited CAR T-cell persistence was also observed in neuroblastoma patients, who received CD8-positive T-cell clones expressing 1st generation CD171-specifc CARs, and in ovarian cancer patients, who received folate receptor (FR-α)-specific CAR T cells. The latter study also highlighted that T-cell homing to solid tumor sites is limited, which at least in preclinical studies can be overcome by genetically modifying T cells with chemokine receptors.; The most promising results (including complete responses) were achieved by Pule et al. with 1st generation CARs directed to GD2, which were expressed in Epstein-Barr virus (EBV)-specific T cells.;
HER2 has been targeted with 2nd and 3rd generation CAR T cells.; One patient developed acute respiratory distress syndrome and died after receiving lymphodepleting chemotherapy, and 1010 T cells expressing a 3rd generation HER2-specific CAR in combination with IL2. In a 2nd study up to 108/m2 T cells expressing a 2nd generation HER2-CAR T cells have been infused. While no overt toxicities were observed, the antitumor activity was limited. Clinical studies with mesothelin-, EGFRvIII-, VEGF-R2-, GD2-, and FAP-specific 2nd or 3rd generation CAR T cells are in progress or will be soon initiated (Table 3).
Table 3
Genetic modification to improve CAR T cells for solid tumors

Genetic modification to enhance CAR T-cell function

Strategies to improve the antitumor activity of CAR T cells include the provision of co-stimulation, the careful selection of T-cell subsets in which to express CARs, and additional genetic modification to enhance CAR T-cell function. We will focus our discussion on additional genetic modifications (Table 3) since the first two strategies are being discussed in detail in other contributions to this themed journal issue.
The solid tumor microenvironment is extremely inhospitable and capable of inducing anergy in CAR T cells. T cells must therefore come armed with countermeasures to thrive in an environment that is replete with immunosuppressive cytokines, regulatory modulators and co-inhibitory receptors.
While inclusion of co-stimulatory signaling domains in the endodomain of CARs can render CAR T cells resistant to inhibitory T cells and/or TGFβ, additional genetic modification strategies are actively being explored to enhance their function. Transgenic expression of cytokines such as IL15;improves CAR T-cell expansion and persistence in vivo, and renders T cells resistant to the inhibitory effects of regulatory T cells (Tregs). Alternatively, transgenic expression of IL12 in CAR T cells reverses the immunosuppressive tumor environment. While there are safety concerns in regards to constitutive IL12 expression, inducible expression systems are available to restrict IL12 production to activated T cells at the tumor site.
Conversely, CAR T cells can be engineered to resist the effects of immunosuppressive cytokines that can inhibit their effector functions. Transforming growth factor (TGF)β is widely used by tumors as an immune evasion strategy, since it limits effector T-cell function and activates regulatory T cells (Tregs). These detrimental effects of TGFβ can be overcome by expressing a dominant negative TGFβ receptor II (DNR);, and this approach is currently being tested in clinical trials. CAR T cells can also be genetically engineered to actively benefit from the inhibitory signals generated by the tumor environment, by expressing chimeric receptors that convert inhibitory signals provided by TGFβ, IL4, or programmed death 1 (PD-1) into stimulatory signals.- Lastly, silencing genes that render T cells susceptible to inhibitory signals in the tumor microenvironment may also improve CAR T-cell function or the transgenic expression of constitutively active signaling molecules.

Targeting the tumor stroma with CAR T cells

Most solid tumors have a stromal compartment that supports tumor growth directly through paracrine secretion of cytokines, growth factors, and provision of nutrients, and contributes to tumor-induced immunosuppression. For example, T cells expressing CARs specific for FAP expressed on cancer associated fibroblasts (CAFs) has potent antitumor effects., To prevent on target/off tumor toxicity,; transient expression of FAP CAR-T cells may be sufficient to weaken the desmoplastic stroma and allow infiltration of tumor specific CAR T cells. Targeting the tumor vasculature with CARs is another attractive strategy ; While the initial CAR ectodomain was based on VEGF to target VEGF receptor 2 (VEGF-R2), more recent studies have used a VEGF-R2-specific scFv, and targeting the tumor vasculature in addition to tumor cells synergized in inducing tumor regression in preclinical models.

Combinatorial CAR T-cell therapy

Combining CAR T cells with other therapies offer the potential to improve antitumor effects. For example, the solid tumor microenvironment abounds in co-inhibitory receptors like cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and PD-1, that are known to limit the efficacy of any directly stimulatory strategy. Systemic administration of antagonists (blocking antibodies) to these co-inhibitory receptors resulted in remarkable response rates even in refractory solid tumors,- and combining blocking antibodies with CAR T cells should boost antitumor effects. Other strategies include epigenetic modifiers that upregulate the expression of tumor associated antigens or the use of targeted therapies that inhibit tumor cells growth, but are not detrimental to T cells.

Conclusions

The ultimate goal of CAR T-cell therapy for solid tumors is to be curative. This requires the development of a potent product that can withstand and thrive in the solid tumor microenvironment. Significant strides have been made towards this end in preclinical studies. While the options are varied, a judicious assessment of a given solid tumor and its microenvironment can help narrow them down to those that would render CAR T cells most successful in inducing a therapeutic response. Clinical trials comparing different genetic modification strategies will be crucial in the future to optimize CAR T cells, transitioning CAR T cells from merely “promising” to being “effective” treatments for solid tumors.

Supplementary Material

Suppl Ref Table 1

Acknowledgments

Support: The authors are supported by NIH grants 1R01CA173750-01 and P01 CA094237, CPRIT grant RP101335, Cookies for Kids’ Cancer, Alex Lemonade Stand Foundation, Dana Foundation, Sidney Kimmel Foundation, and James S McDonnell Foundation.

Footnotes

Conflict of interest: SK is an employee of bluebird bio. The Center for Cell and Gene Therapy has a Research Collaboration with Celgene and bluebird bio. SK and SG have patent applications in the field of T-cell and gene modified T-cell therapy for cancer.

Contributor Information

Sunitha Kakarla, 
Stephen Gottschalk, 

References

1. Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3:388–398. [PMC free article] [PubMed]
2. Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer. 2013;13:525–541. [PubMed]
3. 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. [PMC free article] [PubMed]
4. Sadelain M. Genetic treatment of the haemoglobinopathies: recombinations and new combinations. Br J Haematol. 1997;98:247–253. [PubMed]
5. 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;365:725–733. [PMC free article] [PubMed]
6. Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–1518. [PMC free article] [PubMed]
7. Brentjens RJ, Riviere 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. 2011;118:4817–4828. [PMC free article] [PubMed]
8. 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. 2012;119:2709–2720. [PMC free article] [PubMed]
9. Sampson JH, Heimberger AB, Archer GE, et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28:4722–4729. [PMC free article] [PubMed]
10. Maher J, Wilkie S. CAR mechanics: driving T cells into the MUC of cancer. Cancer Res. 2009;69:4559–4562. [PubMed]
11. Gan HK, Burgess AW, Clayton AH, Scott AM. Targeting of a conformationally exposed, tumor-specific epitope of EGFR as a strategy for cancer therapy. Cancer Res. 2012;72:2924–2930. [PubMed]
12. Coffman KT, Hu M, Carles-Kinch K, et al. Differential EphA2 epitope display on normal versus malignant cells. Cancer Res. 2003;63:7907–7912. [PubMed]
13. Saeki N, Gu J, Yoshida T, Wu X. Prostate stem cell antigen: a Jekyll and Hyde molecule? Clin Cancer Res. 2010;16:3533–3538. [PMC free article] [PubMed]
14. Robbins PF, Morgan RA, Feldman SA, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29:917–924. [PMC free article] [PubMed]
15. Morgan RA, Chinnasamy N, Abate-Daga D, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36:133–151. [PMC free article][PubMed]
16. Willemsen RA, Debets R, Hart E, Hoogenboom HR, Bolhuis RL, Chames P. A phage display selected fab fragment with MHC class I-restricted specificity for MAGE-A1 allows for retargeting of primary human T lymphocytes. Gene Ther. 2001;8:1601–1608. [PubMed]
17. Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2013;31:71–75. [PMC free article] [PubMed]
18. Wilkie S, van Schalkwyk MC, Hobbs S, et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J Clin Immunol. 2012;32:1059–1070. [PubMed]
19. Lanitis E, Poussin M, Klattenhof A, et al. Chimeric Antigen Receptor T Cells with Dissociated Signaling Domains Exhibit Focused Antitumor Activity with Reduced Potential for Toxicity In VivoCancer Immunol Res. 2013 [PMC free article] [PubMed]
20. Grada Z, Hegde M, Byrd T, et al. TanCAR: A Novel Bispecific Chimeric Antigen Receptor for Cancer Immunotherapy. Mol Ther Nucleic Acids. 2013;2:e105. [PMC free article] [PubMed]
21. Urbanska K, Lanitis E, Poussin M, et al. A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor. Cancer Res. 2012;72:1844–1852. [PMC free article][PubMed]
22. Tamada K, Geng D, Sakoda Y, et al. Redirecting gene-modified T cells toward various cancer types using tagged antibodies. Clin Cancer Res. 2012;18:6436–6445. [PubMed]
23. Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A. 2002;99:16168–16173.[PMC free article] [PubMed]
24. Hinrichs CS, Restifo NP. Reassessing target antigens for adoptive T-cell therapy. Nat Biotechnol. 2013;31:999–1008. [PMC free article] [PubMed]
25. 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. 2006;24:e20–e22. [PubMed]
26. Lamers CH, Sleijfer S, van SS, 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;21:904–912. [PMC free article] [PubMed]
27. Park JR, Digiusto DL, Slovak M, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–833. [PubMed]
28. Kershaw MH, Westwood JA, Parker LL, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–6115. [PMC free article][PubMed]
29. 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. 2010 [PMC free article][PubMed]
30. 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. 2002;13:1971–1980. [PubMed]
31. 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. 2008;14:1264–1270. [PMC free article] [PubMed]
32. 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. 2011;118:6050–6056.[PMC free article] [PubMed]
33. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–851. [PMC free article] [PubMed]
34. Ahmed N, Brawley V, Diouf O, et al. T cells redirected against HER2 for the adoptive immunotherapy for HER2-positive osteosarcoma. Cancer Res. 2012;72 Abstract.
35. Gajewski TF, Meng Y, Blank C, et al. Immune resistance orchestrated by the tumor microenvironment. Immunol Rev. 2006;213:131–145. [PubMed]
36. Quintarelli C, Vera JF, Savoldo B, et al. Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes. Blood. 2007;110:2793–2802.[PMC free article] [PubMed]
37. Hoyos V, Savoldo B, Quintarelli C, et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia. 2010;24:1160–1170. [PMC free article] [PubMed]
38. Perna SK, De AB, Pagliara D, et al. Interleukin 15 provides relief to CTLs from regulatory T cell-mediated inhibition: implications for adoptive T cell-based therapies for lymphoma. Clin Cancer Res. 2013;19:106–117. [PMC free article] [PubMed]
39. Kerkar SP, Leonardi AJ, van PN, et al. Collapse of the Tumor Stroma is Triggered by IL-12 Induction of Fas. Mol Ther. 2013;21:1369–1377. [PMC free article] [PubMed]
40. 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. 2011;19:751–759. [PMC free article] [PubMed]
41. Yang L, Pang Y, Moses HL. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010;31:220–227. [PMC free article] [PubMed]
42. Bollard CM, Rossig C, Calonge MJ, et al. Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood. 2002;99:3179–3187. [PubMed]
43. Foster AE, Dotti G, Lu A, et al. Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor. J Immunother. 2008;31:500–505. [PMC free article] [PubMed]
44. Bollard CM, Dotti G, Gottschalk S, et al. Administration of TGF-beta resistant tumor-specific CTL to patienst with EBV-associated HL and NHL. Molecular Therapy. 2012;20:S22.
45. Wilkie S, Burbridge SE, Chiapero-Stanke L, et al. Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4. J Biol Chem. 2010;285:25538–25544.[PMC free article] [PubMed]
46. Leen A, Katari U, Keiman J, Rooney C, Brenner M, Vera J. Improved Expansion and Anti-Tumor Activity of Tumor-Specific CTLs Using a Transgenic Chimeric Cytokine Receptor. Mol Ther. 2011;19:S194.
47. Watanabe N, Anurathapan U, Brenner M, et al. Transgenic Expression of a Novel Immunosuppressive Signal Converter on T Cells. Mol Ther. 2013;22:S153.
48. Ankri C, Shamalov K, Horovitz-Fried M, Mauer S, Cohen CJ. Human T cells engineered to express a programmed death 1/28 costimulatory retargeting molecule display enhanced antitumor activity. J Immunol. 2013;191:4121–4129. [PubMed]
49. Dotti G, Savoldo B, Pule M, et al. Human cytotoxic T lymphocytes with reduced sensitivity to Fas-induced apoptosis. Blood. 2005;105:4677–4684. [PMC free article] [PubMed]
50. Sun J, Dotti G, Huye LE, et al. T cells expressing constitutively active Akt resist multiple tumor-associated inhibitory mechanisms. Mol Ther. 2010;18:2006–2017. [PMC free article] [PubMed]
51. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. [PMC free article] [PubMed]
52. Kakarla S, Chow KK, Mata M, et al. Antitumor Effects of Chimeric Receptor Engineered Human T Cells Directed to Tumor Stroma. Mol Ther. 2013 [PMC free article] [PubMed]
53. Tran E, Chinnasamy D, Yu Z, et al. Immune targeting of fibroblast activation protein triggers recognition of multipotent bone marrow stromal cells and cachexia. J Exp Med. 2013;210:1125–1135.[PMC free article] [PubMed]
54. Roberts EW, Deonarine A, Jones JO, et al. Depletion of stromal cells expressing fibroblast activation protein-alpha from skeletal muscle and bone marrow results in cachexia and anemia. J Exp Med. 2013;210:1137–1151. [PMC free article] [PubMed]
55. Niederman TM, Ghogawala Z, Carter BS, Tompkins HS, Russell MM, Mulligan RC. Antitumor activity of cytotoxic T lymphocytes engineered to target vascular endothelial growth factor receptors. Proc Natl Acad Sci U S A. 2002;99:7009–7014. [PMC free article] [PubMed]
56. 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. 2010;120:3953–3968. [PMC free article] [PubMed]
57. Peggs KS, Quezada SA, Allison JP. Cancer immunotherapy: co-stimulatory agonists and co-inhibitory antagonists. Clin Exp Immunol. 2009;157:9–19. [PMC free article] [PubMed]
58. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. [PMC free article] [PubMed]
59. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. [PMC free article] [PubMed]
60. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;366:2517–2519. [PubMed]
61. Chou J, Voong LN, Mortales CL, et al. Epigenetic modulation to enable antigen-specific T-cell therapy of colorectal cancer. J Immunother. 2012;35:131–141. [PMC free article] [PubMed]
62. Liu C, Peng W, Xu C, et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin Cancer Res. 2013;19:393–403.[PMC free article] [PubMed]
63. 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. 2011;121:1822–1826.[PMC free article] [PubMed]
Logo of mto Molecular Therapy Oncolytics
PMCID: PMC4849432

Chimeric antigen receptor T-cell therapy for solid tumors

Infusion of T cells directed against specific antigens has demonstrated promise in HIV and cancer therapy. Along with immune checkpoint blockade, this approach is triggering a paradigm shift in cancer immunotherapy. Perhaps the most exciting of these approaches has been the use of T cells that have been genetically modified to express chimeric antigen receptor (CAR) genes. CARs are comprised of an extracellular single-chain variable fragment (scFv), which serves as the targeting moiety, a transmembrane spacer, and intracellular signaling/activation domain(s) (Figure 1). The CAR constructs are transfected into T cells, using plasmid transfection, mRNA or via viral vector transduction, to direct them toward tumor-associated antigens (TAAs). CAR structure has evolved significantly from the initial composition involving only the CD3ζ signaling domain, dubbed a “first-generation CAR.” Since then, in an effort to augment T-cell persistence and proliferation, costimulatory endodomains were added, giving rise to second- (e.g., CD3ζ plus 41BB- or CD28-signaling domains) and third-generation (e.g., CD3ζ plus 41BB- and CD28-signaling domains) CARs. CARs have also been constructed in the context of human leukocyte antigen targeting intracellular molecules.
Figure 1
Building blocks of chimeric antigen receptor (CAR) T cell. The single chain (scFv) targeting moiety is taken from the antigen-binding domain of antibodies, fused to the CD3ζ transmembrane and intracellular signaling domains from the T-cell receptor ...
The adoptive transfer of CAR T cells has demonstrated remarkable success in treating blood-borne tumors; prominently, the use of CD19 CARs in leukemias, and indications in patients with lymphoma and myeloma are being explored. A growing number of clinical trials have focused on solid tumors, targeting surface proteins including carcinoembryonic antigen (CEA), the diganglioside GD2, mesothelin, interleukin 13 receptor α (IL13Rα), human epidermal growth factor receptor 2 (HER2), fibroblast activation protein (FAP), and L1 cell adhesion molecule (L1CAM) (reviewed in Gill et al.and Fousek et al.). Unfortunately, the clinical results have been much less encouraging. To date, the two most positive trials reported have used GD2 CARs to target neuroblastoma (3 of 11 patients with complete remissions), and HER2 CARs for sarcoma (4 of 17 patients showing stable disease).
The reason for this is not yet known, but is likely multifactorial. The solid tumor landscape presents unique barriers that are absent in hematological malignancies, and these barriers, either by themselves or in combination with various tumor- and/or host cell-borne factors eventually neutralize CAR activity. Unlike the “liquid tumor” environment of blood malignancies, CAR T cells must successfully traffic to solid tumor sites in spite of potential T-cell chemokine receptor-/tumor-derived chemokine mismatches and successfully infiltrate the stromal elements of solid tumors in order to elicit TAA-specific cytotoxicity, regardless of antigen loss or heterogeneity. Even after successful trafficking and infiltration, T cells must surmount challenges conferred by: (i) an environment characterized by oxidative stress, nutritional depletion, acidic pH, and hypoxia; (ii) the presence of suppressive soluble factors and cytokines; (iii) suppressive immune cells (regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSC), tumor-associated macrophages (TAM) or neutrophils (TAN); and (iv) T-cell-intrinsic negative regulatory mechanisms (e.g., upregulation of cytoplasmic and surface inhibitory receptors) and overexpression of inhibitory molecules. Lastly, the CAR T cells, themselves, may be problematic given their potential immunogenicity and toxicity.
In this mini-review, we discuss some of the key immunosuppressive barriers and other negative elements within solid tumors that ultimately neutralize the function of antitumor T cells, and CAR T cells in particular (Figure 2).
Figure 2
Immunosuppressive tumor microenvironment. This diagram represents a simplified schema of the negative elements that barrage activated chimeric antigen receptor T cells as they navigate through the tumor landscape, thereby inactivating them. These barriers ...

Antigen Specificity and Heterogeneity

The first step in adoptive T-cell therapy is choosing an optimal TAA for CAR T-cell targeting. Ideally, the TAA is highly expressed on the surface of all tumor cells but not on important normal tissues (or at least lowly expressed). However, unlike the success story of CD19 CARs in leukemias, where CD19 is consistently expressed on tumors and only on “dispensable” B cells, specific target antigens on solid tumors have been more difficult to identify. So far, roughly 30 solid tumor antigens are being evaluated for CAR T-cell therapy; these include neoantigens (for instance, mutated sequences), oncofetal or developmental antigens, tumor-selective antigens (i.e., enriched expression of antigens on neoplastic cells, but low basal expression on normal cells), and endogenous tumor-specific antigens; a recent list of CAR targets that are currently being evaluated in clinical trials is available. It should be noted that the scFv avidity to TAA may also be important, and immunoediting and subsequent removal of the most immunogenic epitopes may lead to tumor escape.
Neoantigens on the surface of solid tumors represent an especially attractive target for CAR therapy as their expression is restricted to tumor cells. It is now recognized that most neoantigens are likely to due to tumor-specific mutations and are thus highly individualized (and hence not practical for CAR therapy). However, several neoepitopes that are more generalized have been identified. CAR T cells targeting the mutated EGF receptor is an illustration of this approach; EGFR variant 3 (EGFRvIII) is only expressed on malignant tumor cells (mostly glioblastomas). EGFRvIII CARs have shown promise in treating animal models of glioblastomas, and clinical trials testing the efficacy of EGFRvIII CAR in patients with glioblastomas are underway (NCT02209376NCT01454596). Abnormal glycosylation of extracellular glycoprotein MUC1 is also seen in a large variety of cancers; MUC-1-targeting CAR T cells against MUC1-overexpressing breast cancer xenografts were shown to significantly delay tumor progression. A similar success was reported for CAR T cells targeting MUC16, which is overexpressed in many ovarian carcinomas. CEA is an example of an antigen expressed during developmental growth, but restricted in normal adult tissues and in transformed cells. Evidence of tumor eradication by CEA-CAR T cells in mice has been reported. A recent phase 1 trial using CEA-redirected transgenic T-cell receptor (TCR) T cells, however, showed that while one out of three metastatic colon cancer patients demonstrated an objective response to the therapy as exhibited by regression of metastasis to the lungs and liver, all three patients had to endure transient colitis.Cancers arising from virus transformation may express viral products that are attractive targets for therapy since these products are not displayed on normal tissues; for instance, human papillomavirus (HPV)-transformed ovarian cancers.
Tumor-selective (versus tumor-specific) antigens include targets that are overexpressed on transformed cells but expressed at low levels on normal tissues. This includes mesothelin, a glycoprotein whose overexpression in mesothelioma, ovarian, and pancreatic carcinomas, and low expression on peritoneal, pleural, and pericardial surfaces, has made it an attractive target for CAR therapy., Two mesothelin-specific CARs have been reported; one based on the SS1 antibody—a mouse anti-human scFv which is currently being evaluated in a clinical trial at the University of Pennsylvania (NCT02159716), and another designated P4, a fully human scFv. A fully human scFv targeting mesothelin was recently described by another group, and is currently being tested in a clinical trial (NCT02414269).Treatment using T cells electroporated with the mRNA encoding SS1-CAR, while promising, raised concerns about potential immunogenicity-related toxicity (see below). The notion of targeting multiple antigens (for instance, the expression of 2 scFvs,) or non-tumor cell–related antigens (i.e., a CAR-targeting the stromally-expressed FAP, or the VEGF receptor on tumor endothelium has also been explored.
It should be noted that for any of the targets chosen, the scFv avidity to TAA may also be important. Another issue is that immunoediting and subsequent removal of the most immunogenic epitopes may lead to tumor escape.

Trafficking

Once a CAR that targets appropriate tumor antigen is generated and infused into a patient, an immediate obstacle is the ability of these CAR T cells to successfully target and infiltrate the solid tumor. This process is dependent on the appropriate expression of adhesion receptors on both T cells and the tumor endothelium, and a “match” between the chemokine receptors on the CAR (primarily CXCR3 and CCR5) and the chemokines secreted by the tumors. Unfortunately, there is often a chemokine/chemokine receptor “mismatch”, with tumors producing very small amounts of, for instance, CXCR3 ligands, resulting in inefficient targeting of CXCR3high CD8+ effectors to tumor sites. One approach to overcome this problem is to design CAR T cells that coexpress “better-matched” chemokine receptors. For example, using mesothelioma tumors that make large amounts of CCL2, we and others demonstrated enhanced intratumoral migration of CAR T cells when they coexpressed the appropriate CCR2b transgene, thus leading to subsequent tumor eradication.Similarly, the use of GD2-CAR T cells coexpressing CCR2b exhibited improved trafficking and tumor control compared to GD2-CAR alone. We have also recently found that the genetic inhibition of protein kinase A activation in CAR T cells increased their ability to infiltrate tumors in vivo due to higher baseline expression of CXCR3.
Due to poor trafficking after intravenous injection, local instillation of CARs is also being explored; there are a number of clinical trials that are evaluating the merits of site-specific (i.e., systemic versus regional versus intratumoral) administration of CAR T cells in solid tumors (NCT02498912NCT02414269NCT01818323). One potential limitation is that local instillation is often more technically challenging than simple intravenous administration. Another potential issue is that although site-specific injection of CAR T cells will likely result in higher T-cell levels locally, the ability of these CARs to exit the tumor, enter the blood and then traffic to other tumor sites (which presumably exist in advanced cancer patients) is uncertain. The ongoing studies will help to address these issues.
Several groups have also demonstrated the successful use of oncolytic viruses armed with chemotactic chemokines in attempts to attract CAR T cells to tumor sites. Oncolytic viruses have been shown to successfully and specifically infect tumor cells, and lyse them. The use of oncolytic adenoviral vector expressing CCL5 and GD2-CAR T cells robustly controlled neuroblastoma progression in mice and improved CAR T-cell influx, and similar observations were attained with the use of HER2-CAR T cells loaded with modified oncolytic viruses.

The Hostile Tumor Microenvironment: Physical and Metabolic Barriers

The solid tumor microenvironment presents many problems for CAR T cells. There are purely physical/anatomical barriers, such as stroma that characterizes many types of cancers, and the associated high tissue pressure that prevents extravasation. Countering these barriers by reducing tumor fibroblast numbers using FAP-CAR T cells or by having the CARs secrete an enzyme that degrades matrix have both shown some success in augmenting CAR T-cell function in animal models.
The metabolic landscape within the tumor microenvironment is markedly stressful and inhospitable toward T cells. Prominent hallmarks of the tumor microenvironment include hypoxia and nutrient starvation; under these conditions, elevated lactate generation (leading to acidosis) and the lack of glucose and other metabolites inhibit T-cell proliferation and cytokine production., The lack of nutrients (specifically amino acids such as tryptophan, arginine, and lysine) may also activate the integrated stress response, causing protein translation shutdown or autophagy responses in effector T cells as a means of survival in order to generate an intracellular source of nutrients. For example, the amino acid tryptophan is essential for many biological functions though it cannot be synthesized, and hence must be obtained via dietary means. Tryptophan metabolism as catalyzed by tumor- and MDSC-expressed indolamine-2,3-dioxygenase leads to T-cell anergy and death, and Treg accumulation. In a solid tumor xenograft model of CD19-expressing tumor cells transduced with indolamine-2,3-dioxygenase, Ninomiya and colleagues showed the failure of adoptively transferred CD19 CAR T cells to control progression of indolamine-2,3-dioxygenase-expressing tumors. MDSC may also reduce the bioavailability of the key amino acid arginine (see below). Preliminary work has suggested that manipulation of key cellular regulators of protein synthesis (i.e., the mammalian target of rapamycin) might augment the efficacy of adoptively transferred cells. Sukumar and colleague also showed that inhibiting glycolysis promoted the formation of memory cells, and enhanced antitumor activity.

The Hostile Tumor Microenvironment: Tumor-Derived Soluble Factors and Cytokines

Many studies have reported the presence of immunosuppressive soluble factors in the sera, ascites fluid, and tissue extracts from cancer patients that could inhibit CAR T cells. Prostaglandin E2 (PGE2), a small molecule derivative of arachidonic acid produced by the inducible cyclooxygenase 2 (COX2) enzyme, is generated by both tumor cells and macrophages; many studies have reported PGE2-mediated inhibition of T-cell proliferation, suppression of CD4 help, and subversion of CD8 differentiation. Adenosine, a purine nucleoside seen at high levels during hypoxia, is another potent inhibitor of T-cell proliferation and activity. Both PGE2 and adenosine illicit their immunosuppressive effects via signaling through their own G-coupled receptors which activate protein kinase A in a cyclic AMP-dependent manner. We recently demonstrated that genetic inhibition of protein kinase A activation in CAR T cells can enhance their antitumor efficacy.
Cytokines, implicated in inflammatory responses at tumor sites, can be a double-edged sword, which may bolster or inhibit the antitumor response. One of the most important inhibitory tumor cytokines is TGFβ. In addition to its ability to promote epithelial-to-mesenchymal transition, enhance matrix production, promote metastasis, and skew the immune response toward a Th2 phenotype, TGFβ has direct negative effects on T-cell effector functions. A few approaches have been used to counteract this effect. We previously showed that systemic blockade of TGFβ was efficacious in augmenting adoptive T-cell therapy. To counteract TGFβ effects specifically in T cells, CAR T cells expressing a dominant negative TGFβ receptor have been created. These CAR T cells were resistant to TGFβ suppression and demonstrated augmented efficacy in animal models.
Other inhibitory cytokines include IL10 and IL4; although these have not been directly targeted by alterations in T cells, two groups have constructed chimeric IL4 receptors so that IL4 engagement resulted in signaling that mimicked that of IL2., One of these groups combined this with the use of CAR T cells targeting the tumor-associated antigen MUC1 and showed enhanced efficacy.
Finally, it has also been possible to use introduce activating cytokines to improve the tumor microenvironment milieu to augment CAR function. A few groups have designed CARs or T cells that release the stimulatory cytokine IL12 upon TCR engagement. Although the approach worked extremely well in animal models, a recent clinical trial in which the IL12 gene, driven by an NFAT promoter in adoptively transferred tumor-infiltrating lymphocytes (TILs) resulted in unacceptable toxicity. Finding ways to more tightly control IL12 release or the use of less toxic cytokines (i.e., type 1 interferons) might allow this strategy to proceed in the clinic.

The Hostile Tumor Microenvironment: Immunosuppressive Immune Cells

Within the tumor microenvironment, various suppressive surveilling immune cells, Tregs, MDSC, and TAM/TAN with the so called M2 and N2 phenotype are known to present a barrier against successful antitumor immunity. Although there is extensive literature describing the immunosuppressive nature of these cells, to date, their effects on CAR T-cell therapy has not been extensively examined. One technical factor to consider is that in order to study these cell-cell interactions, mouse CAR T cells must be injected into immunocompetent mice. Given the major differences between the behavior of mouse versus human CAR T cells (e.g., in our experience, mouse CAR T cells are much more sensitive to activation-induced cell death and have a very short persistence compared to human CAR T cells), the relevance of these studies to human CAR T cells is not certain.
MDSC, M2-TAM, and N2-TAN are well-known producers of TGFβ, PGE2, reactive oxygen/nitrogen species, and arginase., As discussed above, all these factors likely blunt the efficacy of CAR T cells. In addition, TAM can express high levels of PDL1, which can interact with PD1 on CAR T cells and inhibit them (see below). MDSC may also recruit Treg cells. On the other hand, TAM and TAN activated in the proper fashion (the so-called M1 or N1 phenotype) can work to eliminate tumor cells.
The role of myeloid cells in CAR therapy is not yet clear. Burga and colleagues found that depletion of GR1+ cells (targeting TAN and MDSC) augmented the ability CEA-CAR T cells to control colorectal cancer liver metastases. In contrast, Spear et al. found in an ovarian cancer model that CARs activated F4/80high TAMs, and enhanced production of nitric oxide by TAMs, leading to tumor lysis. Further studies are needed to more precisely define the role of myeloid cells in CAR efficacy.
CD4+/FOXP3+ Tregs are well-documented suppressors of T-cell activity acting through multiple mechanisms including cell-to-cell contact inhibition, and via soluble factors such as TGFβ, and interleukin 10 (IL10). It has been difficult to study the effects of Tregs on CAR therapy since it is difficult to selectively deplete Tregs. For example, depletion using anti-CD25 antibody will also affect CAR T cells. Nonetheless, some studies have been performed using genetic depletion approaches or adoptive transfer of Tregs with CAR T cells. Zhou et al. studied adoptively transferred cytotoxic T-lymphocytes in a mouse leukemia model and found that antibody blockade of PDL1, combined with genetic depletion (using a diphtheria toxin model) of Tregs, markedly increased efficacy of T-cell adoptive transfer, although depletion of Tregs alone had relatively minor effects. Our lab has recently conducted studies using a selective inhibitor of Tregs and shown augmentation of a mouse CAR T cells targeted to mesothelin (Wang et al., Submitted).
There is some data in humans to suggest an inhibitory effect of Tregs on adoptive T-cell transfer. Perna et al. describe a model in which the efficacy of human GD2-CARs is inhibited by coinjection of human Tregs with IL2. An analysis of four T-cell adoptive therapy clinical trials employing nonmyeloablative chemotherapy with/without total body irradiation before adoptive T-cell transfer revealed that the percentage and number of reconstituting CD4+/FOXP3+ Tregs observed in the peripheral blood was higher in nonresponders than in responders. In addition, the number of administered doses of IL2 was found to be positively associated with peripheral Treg reconstitution. These latter data highlight the complex role of IL2 in CAR therapy. Although IL2 can support CAR T cells in vivo and has been used pre-clinically and in many clinical trials, it also, and perhaps preferentially, activates and induces proliferation of Tregs. Thus, the use of alternative T cells homeostatic cytokines, such as IL7 and IL21, was explored, and shown to enhance CAR efficacy.High IL2 levels with subsequent Treg stimulation may also be an issue in CAR constructs containing the CD28 cytoplasmic domain which produce much higher levels of IL2 than do CARs with the 41BB cytoplasmic domain.

T-Cell-Intrinsic Regulatory Mechanisms

In order to maintain tolerance, T cells express activation-induced surface molecules, such as CTLA4 and PD1, which can have antagonistic effects on the overall antitumor immune response, generally restricting the extent and strength of the immune response upon receptor ligation. The importance of these inhibitory receptors has now been established in multiple clinical trials. Since these receptors are upregulated on infused CAR T cells and even further increased on CAR TILs, a number of groups have shown that blockade of these receptors can augment therapy. For example, using mouse T cells, a combinatorial strategy of HER2-CAR T-cell adoptive transfer and PD1 blockade led to significant tumor regression. In experiments studying human CAR T cells in an immunodeficient animal tumor model, our group showed that PD1 blockade using anti-human antibodies enhanced antitumor effects of human mesothelin-directed CARs. We and Kobold and colleagues showed that it is also possible to reverse the inhibitory effects of PD1 by transducing T cells with a PD1 “switch receptor”; that is, the extracellular domain of PD1 fused to the cytoplasmic domain of an activating receptor like CD28. Antibodies against CTLA4 have also been shown to augment adoptive T-cell transfer.
In addition to surface inhibitory receptors, T cells activate a range of intracellular negative feedback loops after TCR stimulation that work to shut down T-cell activity. Some examples include: (i) enzymes (such as diacylglycerol kinase; (ii) phosphatases (such as SHP1; (ii) ubiquitin ligases (such as Cbl-B); and (iv) transcription factors (such as Ikaros). Augmenting CAR T cells function by reducing the expression or function of these inhibitors is an active area of investigation; for example, CAR T cells lacking expression of diacylglycerol kinase showed markedly increased efficacy.
Another process that can limit CAR function is receptor- or activation-induced cell death. In many cases, this is affected by activation of Fas (CD95) on the T cells through the engagement by Fas ligand (FasL) that is upregulated in most tumor cells, tumor vasculature, and on activated T cells. Engagement of Fas induces T-cell apoptosis, thereby dampening T-cell-mediated immunity. Along these lines, engineering T cells to express higher levels of antiapoptotic proteins was undertaken.

Immunogenicity and Toxicity

Despite the lack of proven efficacy to date, there have been some safety concerns in solid tumor CAR T cells trials that will need to be kept in mind as clinical trials progress. The major toxicity seen in the CAR19 T cells trials has been attributed to severe “cytokine storm” seen in conjunction with rapid T-cell proliferation. It is thought that the infused CAR product causes a widespread, toxic release of proinflammatory cytokines, thus leading to clinical manifestations such as fever, rash, and potentially organ failure. Fortunately (or perhaps unfortunately), this has not yet been observed in trials for solid tumors, likely due to the fact that the degree of T-cell engraftment and proliferation seems to be quite low compared to the leukemia patients. However, as enhanced CARs are developed, and/or as stronger lymphodepletion regimens are employed, this potential toxicity may be observed.
The most feared complication of CAR therapy, a catastrophic and rapid “on target-off tumor” event, has been documented. A fatal event occurred rapidly after infusion with a high affinity HER2-CAR, which was attributed to low-level expression of the antigen on normal endothelium and epithelium.Approaches to avoid this type of event include extensive preclinical toxicology studies, use of “self-limited CARs” that use mRNA rather than lentivirus to transiently express the CAR receptor, and careful dose escalation trial designs. Some groups are also advocating the insertion of suicide genes which can be activated in case of adverse events. Success in preclinical models has been shown with use of the herpes simplex virus thymidine kinase (HSV-TK) gene or an inducible caspase 9 (iCasp9) gene. The activation of these suicide genes leads to the specific and permanent eradication of CAR T cells. Another approach could be to increase the specificity of CARs by requiring the CAR to recognize two antigens to promote activity.,
Finally, the potential immunogenicity of transduced genes must be considered. For example, since the viral gene HSV-TK is immunogenic, the use of iCasp9 seems more attractive as it manipulates the endogenous caspase pathway, and was shown to be very efficient in inducing apoptosis. Phase 1 trials utilizing GD2-CAR T cells with iCasp9 are ongoing (NCT01822652NCT01953900). Another possible problem with immunogenicity relates to the fact that some of the scFvs incorporated in the CARs used in current clinical trials are of murine origin and can thus elicit a human anti-mouse protein immune response. This can be a cellular immune response that eliminates the transduced T cells after 4–6 weeks, but can also result in the generation of anti-murine scFv IgG or even IgE antibodies. In our recent mRNA mesothelin-CAR trial, we observed an anaphylactic reaction when CARs expressing a murine scFv were readministered to a patient after a period of 6 weeks. Because of this, most groups are now using human or humanized scFvs in their CAR constructs.

Conclusions and Future Perspectives

A better understanding of the multiples barriers seen in solid tumors will drive advances in CAR engineering and in clinical trial design. For example, it is currently unclear if aggressive lymphodepletion suggested for TIL therapy will also be needed for CAR T-cell infusion. A number of groups are currently exploring this issue. In preliminary studies from our institution, the use of cyclophosphamide appears to increase blood levels of CARs after infusion, suggesting that some sort of lymphodepletion may be needed in solid tumor therapy.
Some approaches to overcome solid tumor barriers were discussed above, however many other strategies are being tested. To mention just a few, the use of alternative cytoplasmic activation domains, such as ICOS, 41BB, OX40, or CD27 are being explored., Even more radical design changes are also being evaluated. Wang et al. have fused a scFv for antigen recognition to the transmembrane and cytoplasmic domains of KIR2DS2, a stimulatory killer immunoglobulin-like receptor (KIR). This KIR-based CAR (KIR-CAR), when fused to the adaptor DAP12, proliferated in an antigen-specific manner, and demonstrated enhanced effector function.
The compelling success of CAR therapy in hematologic malignancies is propelling the development of CARs that can show similar efficacy in solid tumors. The ability to genetically manipulate infused CAR T cells provides almost limitless opportunities for additional changes and improvements, and thus provides strong hope for future success.

Notes

The authors declare no conflict of interest.

References

  • Topalian, SL, Drake, CG and Pardoll, DM (2015). Immune checkpoint blockade: a common denominator approach to cancer therapyCancer Cell 27: 450–461. [PMC free article] [PubMed]
  • Willemsen, RA, Debets, R, Hart, E, Hoogenboom, HR, Bolhuis, RL and Chames, P (2001). A phage display selected fab fragment with MHC class I-restricted specificity for MAGE-A1 allows for retargeting of primary human T lymphocytesGene Ther 8: 1601–1608. [PubMed]
  • Gill, S, Maus, MV and Porter, DL (2015). Chimeric antigen receptor T cell therapy: 25 years in the makingBlood Rev (Epub ahead of print). [PubMed]
  • Fousek, K and Ahmed, N (2015). The evolution of T-cell therapies for solid malignanciesClin Cancer Res 21: 3384–3392. [PMC free article] [PubMed]
  • Louis, CU, Savoldo, B, Dotti, G, Pule, M, Yvon, E, Myers, GD et al. (2011). Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastomaBlood 118: 6050–6056. [PMC free article] [PubMed]
  • Ahmed, N, Brawley, VS, Hegde, M, Robertson, C, Ghazi, A, Gerken, C et al. (2015). Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcomaJ Clin Oncol 33: 1688–1696. [PMC free article] [PubMed]
  • Restifo, NP, Dudley, ME and Rosenberg, SA (2012). Adoptive immunotherapy for cancer: harnessing the T cell responseNat Rev Immunol 12: 269–281. [PubMed]
  • Johnson, LA, Scholler, J, Ohkuri, T, Kosaka, A, Patel, PR, McGettigan, SE et al. (2015). Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastomaSci Transl Med 7: 275ra22. [PMC free article] [PubMed]
  • Morgan, RA, Johnson, LA, Davis, JL, Zheng, Z, Woolard, KD, Reap, EA et al. (2012). Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for gliomaHum Gene Ther 23: 1043–1053. [PMC free article] [PubMed]
  • Wilkie, S, Picco, G, Foster, J, Davies, DM, Julien, S, Cooper, L et al. (2008). Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptorJ Immunol 180: 4901–4909. [PubMed]
  • Chekmasova, AA, Rao, TD, Nikhamin, Y, Park, KJ, Levine, DA, Spriggs, DR et al. (2010). Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigenClin Cancer Res 16: 3594–3606. [PMC free article] [PubMed]
  • Chmielewski, M, Hahn, O, Rappl, G, Nowak, M, Schmidt-Wolf, IH, Hombach, AA et al. (2012). T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in miceGastroenterology 143: 1095–107.e2. [PubMed]
  • Parkhurst, MR, Yang, JC, Langan, RC, Dudley, ME, Nathan, DA, Feldman, SA et al. (2011). T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitisMol Ther 19: 620–626. [PMC free article] [PubMed]
  • Kenter, GG, Welters, MJ, Valentijn, AR, Lowik, MJ, Berends-van der Meer, DM, Vloon, AP et al. (2009). Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasiaN Engl J Med 361: 1838–1847. [PubMed]
  • Morello, A, Sadelain, M and Adusumilli, PS (2016). Mesothelin-targeted CARs: driving T cells to solid tumorsCancer Discov 6: 133–146. [PMC free article] [PubMed]
  • Pastan, I and Hassan, R (2014). Discovery of mesothelin and exploiting it as a target for immunotherapyCancer Res 74: 2907–2912. [PMC free article] [PubMed]
  • Chowdhury, PS, Viner, JL, Beers, R and Pastan, I (1998). Isolation of a high-affinity stable single-chain Fv specific for mesothelin from DNA-immunized mice by phage display and construction of a recombinant immunotoxin with anti-tumor activityProc Natl Acad Sci USA95: 669–674. [PMC free article] [PubMed]
  • Lanitis, E, Poussin, M, Hagemann, IS, Coukos, G, Sandaltzopoulos, R, Scholler, N et al. (2012). Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptorMol Ther 20: 633–643. [PMC free article] [PubMed]
  • Adusumilli, PS, Cherkassky, L, Villena-Vargas, J, Colovos, C, Servais, E, Plotkin, J et al. (2014). Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunitySci Transl Med 6: 261ra151. [PMC free article] [PubMed]
  • Wilkie, S, van Schalkwyk, MC, Hobbs, S, Davies, DM, van der Stegen, SJ, Pereira, AC et al. (2012). Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signalingJ Clin Immunol 32: 1059–1070. [PubMed]
  • Wang, LC, Lo, A, Scholler, J, Sun, J, Majumdar, RS, Kapoor, V et al. (2014). Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicityCancer Immunol Res 2: 154–166. [PMC free article] [PubMed]
  • Chinnasamy, D, Tran, E, Yu, Z, Morgan, RA, Restifo, NP and Rosenberg, SA (2013). Simultaneous targeting of tumor antigens and the tumor vasculature using T lymphocyte transfer synergize to induce regression of established tumors in miceCancer Res 73: 3371–3380. [PMC free article] [PubMed]
  • Harlin, H, Meng, Y, Peterson, AC, Zha, Y, Tretiakova, M, Slingluff, C et al. (2009). Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitmentCancer Res 69: 3077–3085. [PMC free article] [PubMed]
  • Moon, EK, Carpenito, C, Sun, J, Wang, LC, Kapoor, V, Predina, J et al. (2011). Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptorClin Cancer Res 17: 4719–4730. [PMC free article] [PubMed]
  • Craddock, JA, Lu, A, Bear, A, Pule, M, Brenner, MK, Rooney, CM et al. (2010). Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2bJ Immunother 33: 780–788. [PMC free article] [PubMed]
  • Newick, K, O’Brien, S, Sun, J, Kapoor, V, Maceyko, S, Lo, A et al. (2016). Augmentation of CAR T cell trafficking and antitumor efficact by blocking protein kinase A (PKA) localizationCancer Immunol Res. In press. [PMC free article] [PubMed]
  • Nishio, N, Diaconu, I, Liu, H, Cerullo, V, Caruana, I, Hoyos, V et al. (2014). Armed oncolytic virus enhances immune functions of chimeric antigen receptor-modified T cells in solid tumorsCancer Res 74: 5195–5205. [PMC free article] [PubMed]
  • VanSeggelen, H, Hammill, JA, Dvorkin-Gheva, A, Tantalo, DG, Kwiecien, JM, Denisova, GF et al. (2015). T cells engineered with chimeric antigen receptors targeting NKG2D ligands display lethal toxicity in miceMol Ther 23: 1600–1610. [PMC free article] [PubMed]
  • Caruana, I, Savoldo, B, Hoyos, V, Weber, G, Liu, H, Kim, ES et al. (2015). Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytesNat Med 21: 524–529. [PMC free article] [PubMed]
  • Jacobs, SR, Herman, CE, Maciver, NJ, Wofford, JA, Wieman, HL, Hammen, JJ et al. (2008). Glucose uptake is limiting in T cell activation and requires CD28-mediated Akt-dependent and independent pathwaysJ Immunol 180: 4476–4486. [PMC free article] [PubMed]
  • Fischer, K, Hoffmann, P, Voelkl, S, Meidenbauer, N, Ammer, J, Edinger, M et al. (2007). Inhibitory effect of tumor cell-derived lactic acid on human T cellsBlood 109: 3812–3819.[PubMed]
  • Howie, D, Waldmann, H and Cobbold, S (2014). Nutrient sensing via mTOR in T cells maintains a tolerogenic microenvironmentFront Immunol 5: 409. [PMC free article] [PubMed]
  • Ninomiya, S, Narala, N, Huye, L, Yagyu, S, Savoldo, B, Dotti, G et al. (2015). Tumor indoleamine 2,3-dioxygenase (IDO) inhibits CD19-CAR T cells and is downregulated by lymphodepleting drugsBlood 125: 3905–3916. [PMC free article] [PubMed]
  • Veliça, P, Zech, M, Henson, S, Holler, A, Manzo, T, Pike, R et al. (2015). Genetic regulation of fate decisions in therapeutic T cells to enhance tumor protection and memory formationCancer Res 75: 2641–2652. [PubMed]
  • Sukumar, M, Liu, J, Ji, Y, Subramanian, M, Crompton, JG, Yu, Z et al. (2013). Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor functionJ Clin Invest 123: 4479–4488. [PMC free article] [PubMed]
  • Goodwin, JS, Bankhurst, AD and Messner, RP (1977). Suppression of human T-cell mitogenesis by prostaglandin. Existence of a prostaglandin-producing suppressor cellJ Exp Med 146: 1719–1734. [PMC free article] [PubMed]
  • Su, Y, Huang, X, Raskovalova, T, Zacharia, L, Lokshin, A, Jackson, E et al. (2008). Cooperation of adenosine and prostaglandin E2 (PGE2) in amplification of cAMP-PKA signaling and immunosuppressionCancer Immunol Immunother 57: 1611–1623. [PubMed]
  • Oh, SA and Li, MO (2013). TGF-ß: guardian of T cell functionJ Immunol 191: 3973–3979. [PMC free article] [PubMed]
  • Massagué, J (2008). TGFbeta in CancerCell 134: 215–230. [PMC free article] [PubMed]
  • Wallace, A, Kapoor, V, Sun, J, Mrass, P, Weninger, W, Heitjan, DF et al. (2008). Transforming growth factor-beta receptor blockade augments the effectiveness of adoptive T-cell therapy of established solid cancersClin Cancer Res 14: 3966–3974. [PMC free article] [PubMed]
  • Bollard, CM, Rössig, C, Calonge, MJ, Huls, MH, Wagner, HJ, Massague, J et al. (2002). Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunityBlood 99: 3179–3187. [PubMed]
  • Leen, AM, Sukumaran, S, Watanabe, N, Mohammed, S, Keirnan, J, Yanagisawa, R et al. (2014). Reversal of tumor immune inhibition using a chimeric cytokine receptorMol Ther 22: 1211–1220. [PMC free article] [PubMed]
  • Wilkie, S, Burbridge, SE, Chiapero-Stanke, L, Pereira, AC, Cleary, S, van der Stegen, SJ et al. (2010). Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4J Biol Chem 285: 25538–25544. [PMC free article] [PubMed]
  • Chmielewski, M, Hombach, AA and Abken, H (2014). Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stromaImmunol Rev 257: 83–90. [PubMed]
  • Zhang, L, Morgan, RA, Beane, JD, Zheng, Z, Dudley, ME, Kassim, SH et al. (2015). Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanomaClin Cancer Res 21: 2278–2288. [PMC free article] [PubMed]
  • Gabrilovich, DI and Nagaraj, S (2009). Myeloid-derived suppressor cells as regulators of the immune systemNat Rev Immunol 9: 162–174. [PMC free article] [PubMed]
  • Mussai, F, Egan, S, Hunter, S, Webber, H, Fisher, J, Wheat, R et al. (2015). Neuroblastoma Arginase Activity Creates an Immunosuppressive Microenvironment That Impairs Autologous and Engineered ImmunityCancer Res 75: 3043–3053. [PMC free article] [PubMed]
  • Burga, RA, Thorn, M, Point, GR, Guha, P, Nguyen, CT, Licata, LA et al. (2015). Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-TCancer Immunol Immunother 64: 817–829. [PMC free article][PubMed]
  • Spear, P, Barber, A, Rynda-Apple, A and Sentman, CL (2012). Chimeric antigen receptor T cells shape myeloid cell function within the tumor microenvironment through IFN-? and GM-CSFJ Immunol 188: 6389–6398. [PMC free article] [PubMed]
  • Sakaguchi, S, Miyara, M, Costantino, CM and Hafler, DA (2010). FOXP3+ regulatory T cells in the human immune systemNat Rev Immunol 10: 490–500. [PubMed]
  • Zhou, Q, Munger, ME, Highfill, SL, Tolar, J, Weigel, BJ, Riddle, M et al. (2010). Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemiaBlood 116: 2484–2493. [PMC free article] [PubMed]
  • Liu, Y, Wang, L, Predina, J, Han, R, Beier, UH, Wang, LC et al. (2013). Inhibition of p300 impairs Foxp3? T regulatory cell function and promotes antitumor immunityNat Med 19: 1173–1177. [PMC free article] [PubMed]
  • Perna, SK, Pagliara, D, Mahendravada, A, Liu, H, Brenner, MK, Savoldo, B et al. (2014). Interleukin-7 mediates selective expansion of tumor-redirected cytotoxic T lymphocytes (CTLs) without enhancement of regulatory T-cell inhibitionClin Cancer Res 20: 131–139. [PMC free article] [PubMed]
  • Yao, X, Ahmadzadeh, M, Lu, YC, Liewehr, DJ, Dudley, ME, Liu, F et al. (2012). Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancerBlood 119: 5688–5696. [PMC free article] [PubMed]
  • Kofler, DM, Chmielewski, M, Rappl, G, Hombach, A, Riet, T, Schmidt, A et al. (2011). CD28 costimulation Impairs the efficacy of a redirected t-cell antitumor attack in the presence of regulatory T cells which can be overcome by preventing Lck activationMol Ther 19: 760–767. [PMC free article] [PubMed]
  • Markley, JC and Sadelain, M (2010). IL-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in immunodeficient miceBlood 115: 3508–3519. [PMC free article] [PubMed]
  • Finney, HM, Akbar, AN and Lawson, AD (2004). Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chainJ Immunol 172: 104–113. [PubMed]
  • Moon, EK, Wang, LC, Dolfi, DV, Wilson, CB, Ranganathan, R, Sun, J et al. (2014). Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumorsClin Cancer Res 20: 4262–4273. [PMC free article] [PubMed]
  • John, LB, Devaud, C, Duong, CP, Yong, CS, Beavis, PA, Haynes, NM et al. (2013). Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cellsClin Cancer Res 19: 5636–5646. [PubMed]
  • Moon, EK, Ranganathan, R, Eruslanov, E, Kim, S, Newick, K, O’Brien, S et al. (2016). Blockade of programmed death 1 augments the ability of human T cells engineered to target NY-ESO-1 to control tumor growth after adoptive transferClin Cancer Res 22: 436–447. [PMC free article][PubMed]
  • Kobold, S, Grassmann, S, Chaloupka, M, Lampert, C, Wenk, S, Kraus, F et al. (2015). Impact of a new fusion receptor on PD-1-mediated immunosuppression in adoptive T cell therapyJ Natl Cancer Inst 107:djv146 . [PMC free article] [PubMed]
  • Mahvi, DA, Meyers, JV, Tatar, AJ, Contreras, A, Suresh, M, Leverson, GE et al. (2015). Ctla-4 blockade plus adoptive T-cell transfer promotes optimal melanoma immunity in miceJ Immunother 38: 54–61. [PMC free article] [PubMed]
  • Wang, LC, Riese, MJ, Moon, EK and Albelda, SM (2013). Overcoming intrinsic inhibitory pathways to augment the antineoplastic activity of adoptively transferred T cells: Re-tuning your CAR before hitting a rocky roadOncoimmunology 2: e26492. [PMC free article] [PubMed]
  • Riese, MJ, Wang, LC, Moon, EK, Joshi, RP, Ranganathan, A, June, CH et al. (2013). Enhanced effector responses in activated CD8+ T cells deficient in diacylglycerol kinasesCancer Res 73: 3566–3577. [PMC free article] [PubMed]
  • Charo, J, Finkelstein, SE, Grewal, N, Restifo, NP, Robbins, PF and Rosenberg, SA (2005). Bcl-2 overexpression enhances tumor-specific T-cell survivalCancer Res 65: 2001–2008. [PMC free article] [PubMed]
  • Ramos, CA, Savoldo, B and Dotti, G (2014). CD19-CAR trialsCancer J 20: 112–118. [PMC free article] [PubMed]
  • Lee, DW, Gardner, R, Porter, DL, Louis, CU, Ahmed, N, Jensen, M et al. (2014). Current concepts in the diagnosis and management of cytokine release syndromeBlood 124: 188–195. [PMC free article] [PubMed]
  • Morgan, RA, Yang, JC, Kitano, M, Dudley, ME, Laurencot, CM and Rosenberg, SA (2010). Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2Mol Ther 18: 843–851. [PMC free article][PubMed]
  • Lanitis, E, Poussin, M, Klattenhoff, AW, Song, D, Sandaltzopoulos, R, June, CH et al. (2013). Chimeric antigen receptor T cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivoCancer Immunol Res 1: 43–53. [PMC free article] [PubMed]
  • Kloss, CC, Condomines, M, Cartellieri, M, Bachmann, M and Sadelain, M (2013). Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cellsNat Biotechnol 31: 71–75. [PMC free article] [PubMed]
  • Tey, SK, Dotti, G, Rooney, CM, Heslop, HE and Brenner, MK (2007). Inducible caspase 9 suicide gene to improve the safety of allodepleted T cells after haploidentical stem cell transplantationBiol Blood Marrow Transplant 13: 913–924. [PMC free article] [PubMed]
  • Beatty, GL, Haas, AR, Maus, MV, Torigian, DA, Soulen, MC, Plesa, G et al. (2014). Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignanciesCancer Immunol Res 2: 112–120. [PMC free article] [PubMed]
  • Guedan, S, Chen, X, Madar, A, Carpenito, C, McGettigan, SE, Frigault, MJ et al. (2014). ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cellsBlood 124: 1070–1080. [PMC free article] [PubMed]
  • Song, DG, Ye, Q, Poussin, M, Harms, GM, Figini, M and Powell, DJ Jr (2012). CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivoBlood 119: 696–706. [PubMed]
  • Wang, E, Wang, LC, Tsai, CY, Bhoj, V, Gershenson, Z, Moon, E et al. (2015). Generation of potent T-cell immunotherapy for cancer using DAP12-based, multichain, chimeric immunoreceptorsCancer Immunol Res 3: 815–826. [PMC free article] [PubMed]

Articles from Molecular Therapy Oncolytics are provided here courtesy of American Society of Gene & Cell Therapy

CAR T cell therapy for solid tumors

K Newick, S O'Brien, E Moon… - Annual review of …, 2017 - annualreviews.org
... Another potential issue is that although site-specific injection of CAR T cells will likely result in
higher T cell levels locally, the ability of these CARs to exit the tumor, enter the blood, and then
traffic to other tumor sites (which presumably exist in advanced cancer ...

Targeting of Aberrant αvβ6 Integrin Expression in Solid Tumors Using Chimeric Antigen Receptor-Engineered T Cells

LM Whilding, AC Parente-Pereira, T Zabinski… - Molecular Therapy, 2017 - Elsevier
... A second αvβ6-specific CAR-targeting moiety was engineered by placing the B12 peptide
downstream of a ... (E) The SFG retroviral vector was used to express CARs in human ... IL-4-responsive
4αβ chimeric cytokine receptor was achieved using a Thosea Asigna (T)2A ribosomal ...

[HTML] Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition

J Ren, X Liu, C Fang, S Jiang, CH June, Y Zhao - Clinical Cancer Research, 2017 - AACR
Skip to main content. AACR Publications: Cancer Discovery; Cancer Epidemiology, Biomarkers
& Prevention; Cancer Immunology Research; Cancer Prevention Research; Cancer Research;
Clinical Cancer Research; Molecular Cancer Research; Molecular Cancer Therapeutics ...

Targeting the tumor and its associated stroma: One and one can make three in adoptive T cell therapy of solid tumors

A Mondino, G Vella, L Icardi - Cytokine & Growth Factor Reviews, 2017 - Elsevier
... Attempts are now directed towards the identification of tumor-specific mutated antigens,
and corresponding TCR [21] ; [22]. Chimeric antigen receptors (CAR) are also being
exploited to redirect autologous T cells to tumor surface antigens. ...

… -label study of GSK3359609, an ICOS agonist antibody, administered alone and in combination with pembrolizumab in patients with selected, advanced solid tumors

E Angevin, TM Bauer, CE Ellis, H Gan, R Hall… - 2017 - AACR
Skip to main content. AACR Publications: Cancer Discovery; Cancer Epidemiology, Biomarkers
& Prevention; Cancer Immunology Research; Cancer Prevention Research; Cancer Research;
Clinical Cancer Research; Molecular Cancer Research; Molecular Cancer Therapeutics ...

Abstract CT118: T4 immunotherapy of head and neck squamous cell carcinoma using pan-ErbB targeted CAR T-cells

…, S van der Stegen, DM Davies, T Guerrero-Urbano… - 2017 - AACR
Skip to main content. AACR Publications: Cancer Discovery; Cancer Epidemiology, Biomarkers
& Prevention; Cancer Immunology Research; Cancer Prevention Research; Cancer Research;
Clinical Cancer Research; Molecular Cancer Research; Molecular Cancer Therapeutics ...

Longitudinal and quantitative imaging of the localization, expansion, and contraction of tumor targeted adoptively transferred T cells

Y Vedvyas, E Shevlin, M Zaman, IM Min, MM Jin - 2017 - AACR
Skip to main content. AACR Publications: Cancer Discovery; Cancer Epidemiology, Biomarkers
& Prevention; Cancer Immunology Research; Cancer Prevention Research; Cancer Research;
Clinical Cancer Research; Molecular Cancer Research; Molecular Cancer Therapeutics ...

Possible Compartmental Cytokine Release Syndrome in a Patient With Recurrent Ovarian Cancer After Treatment With Mesothelin-targeted CAR-T Cells

JL Tanyi, C Stashwick, G Plesa… - Journal of …, 2017 - journals.lww.com
You may be trying to access this site from a secured browser on the server. Please
enable scripts and reload this page. Account: Saved Searches; ... your express consent.
For more information, please refer to our Privacy Policy. ...

[HTML] CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells

LJ Rupp, K Schumann, KT Roybal, RE Gate… - Scientific Reports, 2017 - nature.com
... impairs CAR T cell signaling or function, it might be possible to engineer CARs with additional
or alternative costimulatory domains that are resistant to PD-L1 ... Given that CAR T cells with different
costimulatory domains (such as those derived from CD28, 4-1BB, or ...


http://ift.tt/2u3YOPa

Δεν υπάρχουν σχόλια:

Δημοσίευση σχολίου

Δημοφιλείς αναρτήσεις