Skip to main content
Erschienen in: Molecular Cancer 1/2018

Open Access 01.12.2018 | Review

Advances on chimeric antigen receptor-modified T-cell therapy for oncotherapy

verfasst von: Yanyu Pang, Xiaoyang Hou, Chunsheng Yang, Yanqun Liu, Guan Jiang

Erschienen in: Molecular Cancer | Ausgabe 1/2018

Abstract

Tumor treatment is still complicated in the field of medicine. Tumor immunotherapy has been the most interesting research field in cancer therapy. Application of chimeric antigen receptor T (CAR-T) cell therapy has recently achieved excellent clinical outcome in patients, especially those with CD19-positive hematologic malignancies. This phenomenon has induced intense interest to develop CAR-T cell therapy for cancer, especially for solid tumors. However, the performance of CAR-T cell treatment in solid tumor is not as satisfactory as that in hematologic disease. Clinical studies on some neoplasms, such as glioblastoma, ovarian cancer, and cholangiocarcinoma, have achieved desirable outcome. This review describes the history and evolution of CAR-T, generalizes the structure and preparation of CAR-T, and summarizes the latest advances on CAR-T cell therapy in different tumor types. The last section presents the current challenges and prospects of CAR-T application to provide guidance for subsequent research.
Hinweise
Yanyu Pang and Xiaoyang Hou contributed equally to this work.
Abkürzungen
ALL
Acute lymphoblastic leukemia
CAR
Chimeric antigen receptor
CCA
Cholangiocarcinoma
CEA
Carcinoembryonic antigen
CR
Complete remissions
CRC
Colorectal cancer
CRS
Cytokine-release syndrome
EGFR
Epidermal growth factor receptor
EOC
Epithelial ovarian cancer
EphA2
Erythropoietin-producing hepatocellular carcinoma A2
FDA
Food and Drug Administration
FRα
Folate receptor-α
GBM
Glioblastoma
IL13Rα2
IL-13 receptor α2
L1-CAM
L1-cell adhesion molecule
MHC
Major histocompatibility complex
MM
Multiple myeloma
MPM
Malignant pleural mesothelioma
mRCC
Metastatic renal cell carcinoma
MUC1
Mucin 1
NHLs
non-Hodgkin’s lymphomas
NSCLC
Non-small cell lung cancer
PR
Partial response
PSMA
Prostate-specific membrane antigen
scFv
Single-chain variable region domain
SR
Stable diseases
SVC
Seminal vesicle cancer
TAAs
Tumor-associated antigen
TAG72
Tumor-associated glycoprotein 72
TCR
T cell receptor
TILs
Tumor-infiltrating lymphocytes

Background

Despite the rapid development in medical science and the emergence of new medical technology, tumor therapy is still an intractable problem. Conventional therapies, such as surgery, chemotherapy, and radiotherapy, may provide short-term benefits but have annoying side effects due to their invasiveness and biotoxicity [1, 2]. Furthermore, multidrug resistance for chemotherapy and multiple toxicities of radiotherapy limit their curative effects [3, 4]. Therefore, new and effective treatments must be developed. Typical immunotherapy, including the use of tumor-infiltrating lymphocytes (TILs), T cell receptor (TCR)-engineered T cells, and chimeric antigen receptor (CAR) -modified T cells, has harnessed the immune system against cancer and emerged as a promising treatment modality for human malignancies [57]. TILs are cultured from fragments of resected tumors and have produced encouraging results in the therapy of metastatic melanoma [8] but are limited in other solid tumors due to the difficulty in isolation and expansion in vitro [9]. TCR T cell therapy is restricted to major histocompatibility complex (MHC)-expressing antigens [10]. Alternatively, CAR-T cell-based immunotherapy is independent of MHC [11, 12] and has achieved spectacular success in treating cancers, especially B-cell hematologic malignancies [1315]. CARs are recombinant receptors containing an extracellular antigen recognition domain, a transmembrane domain, and a cytoplasmic signaling domain (such as CD3ζ, CD28, and 4-1BB). Therefore, T cells expressing CAR can recognize a wide range of cell surface antigens, including glycolipids, carbohydrates, and proteins [16], and can attack malignant cells expressing these antigens through the activation of cytoplasmic costimulation [12]. On July 1, 2014, the US Food and Drug Administration (FDA) granted the breakthrough therapy designation to CTL019, which is the CD19-directed CAR-T cell therapy designed by the University of Pennsylvania [17]. In 2017, FDA successively approved two drugs, namely, tisagenlecleucel (CTL019, Novartis) for the treatment of children and young adults with relapsing/refractory acute lymphoblastic leukemia (r/r ALL) and axicabtagene ciloleucel (KTE-C19, Kite Pharma) for the treatment of non-Hodgkin’s lymphomas (NHLs) [18, 19]. CAR-T cell therapy also has some effects in other diseases, such as in non-small cell lung cancer (NSCLC), malignant pleural mesothelioma (MPM), metastatic renal cell carcinoma (mRCC), and glioblastoma (GBM) [2023]. Although the therapeutic efficacy of CAR-T cell in these solid tumors is less effective than that in hematologic diseases, the successful trials achieved by CAR-T cells provide a concrete platform for its further development in solid tumors. In this review, we analyze the reasons why CAR-T cell therapy reaches its limits when targeting solid tumors, conclude the applications of CAR-T cell therapy in different tumors, and discuss the future perspectives on CAR-T cell therapy in cancer treatment.

CAR-T profile

History and evolution

In 1989, as the beginning of CAR-T cell, Eshhar and colleagues first generated chimeric TCR genes that can be functionally expressed in T cells and endowed the recipient T cell with antibody-type specificity to recognize and respond to the antigen in a non-MHC-restricted manner [12]. In 1993, to achieve the advantages of antibody specificity and T-cell cytotoxic activity, Eshhar combined a single-chain variable region domain (scFv) of an antibody molecule with the constant region domain of the TCR, which is usually the ζ chain of the TCR/CD3 complex [24], to construct a chimeric receptor gene and subsequently induce the T cells to express this gene by generating chimeric scFvRζ T cells [25], which are later called “first-generation CARs” that unfortunately showed limited clinical benefit because of failure in directing T cell expansion upon repeated exposure to the antigen [26] (Table 1). Hence, a co-stimulatory signaling domain–CD28 or 41BB–was added in between scFv and CD3ζ chain to form the “second-generation CARs.” This domain sustainably activates the T cell to augment cytokine secretion and amplify T cell proliferation; thus, the T cells can expand upon repeated antigen exposure and show significant clinical responses [27, 28]. “Third-generation CARs” were formed by incorporating two or more costimulatory domains, usually CD28 and 41BB (CD137) or OX40 (CD134), into the same CAR. However, whether it has better clinical effect than second-generation CAR-T remains unclear [29]. In general, third-generation CARs enhance the expansion and persistence of CAR-T cells after tumor challenge [30, 31]. Notwithstanding, Haso et al. reported that in most vitro cases of anti-CD22 CARs for B-cell ALL, second-generation CAR was superior over third-generation CAR [32]. Moreover, a clinical trial of CEA CAR-T therapy on patients with carcino-embryonic antigen (CEA)-positive colorectal cancer (CRC) carried out by Zhang et al. showed that the third generation of CAR with CD28 and CD137 signaling does not show better performance than the second generation with CD28 signaling [33]. The significant phenotypic heterogeneity of solid tumors makes it difficult for CAR to recognize cancer cells. To circumvent these barriers in solid tumor lesions, Markus Chmielewski et al. developed the “fourth-generation CAR” (TRUCKs, T cells redirected for universal cytokine killing) that include the costimulatory domain and the CAR-inducible interleukin-12 (iIL-12) cassette. When CAR binds to target antigen, it activates T cell signaling; iIL-12 cassette then secretes pro-inflammatory IL-12, which can accumulate in the targeted tissue and thus recruit a second wave of immune cells (NK cells, macrophages) to initiate an attack toward those that would normally escape cancer cells due to the lack of CAR-recognized target and invisibility to CAR-T cells [34, 35].
Table 1
Summary and comparison of four generations of CAR-T therapy
CAR generations
Signal domain
Target antigen
Associated diseases
Profile
References
1st
 
CD3ζ
TAG72
Metastatic colorectal cancer
Limited persistence
[84]
 
CD3ζ
FRα
Ovarian cancer
Limited persistence
[26]
 
CD3ζ
L1-CAM
Metastatic neuroblastoma
Limited persistence
[85]
2nd
 
CD3ζ + CD28/CD137 (41BB)
CD19
B cell lymphomas
Enhanced expansion, persistence and anti-tumor effect
[28, 40, 86, 87]
 
CD3ζ + 41BB(CD137)
IL13Rα2
GBM
Improved anti-tumor activity and T cell persistence
[22]
 
CD3ζ + 41BB (CD137)
FRα
Ovarian cancer
Augmented cytokine secretion and proliferation
[88]
3rd
 
CD3ζ + CD28 + 41BB(CD137)
CD19
ALL
Superior activation and proliferation capacity
[89]
 
CD3ζ + CD28 + 41BB(CD137)
PMSA
Promoted cytokine release, T-cell survival and tumor elimination
[90]
 
CD3ζ + CD28 + CD137 (41BB)
Mesothelin
Mesothelioma
Prolonged persistence
[30]
 
CD3ζ + CD28 + 41BB(CD137)
CD22
ALL
Inferior antileukemic activity
[32]
4th
 
CD3ζ + iIL-12+ co-stimulator
CEA
CEA+ tumors
Improved antitumor efficacy
[35]
TAG72 tumor-associated glycoprotein 72, CEA carcinoembryonic antigen, IL13Rα2 IL-13 receptor α2, FRα folate receptor-α, L1-CAM L1-cell adhesion molecule, PSMA prostate-specific membrane antigen

Structure

CARs are engineered receptors that possess both antigen-binding and T-cell-activating functions. Based on the location of the CAR in the membrane of the T cell, CAR can be divided into three main distinct modules (Fig. 1), that is an extracellular antigen-binding domain, followed by a space region, a transmembrane domain, and the intracellular signaling domain. The antigen-binding moiety, most commonly derived from variable regions of immunoglobulin, is composed of VH and VL chains that are joined up by a linker to form the so-called “scFv” [12, 25]. The segment interposing between the scFv and the transmembrane domain is a “spacer domain,” that is commonly the constant IgG1 hinge-CH2–CH3 Fc domain [36]. In some cases, the space domain and the transmembrane domain are derived from CD8 [37]. The intracellular signaling domains mediating T cell activation include a CD3ζ co-receptor signaling domain derived from C-region of the TCR α and β chains [12] and one or more costimulatory domains.

Preparation

The manufacturing processes of CAR-T cells are complex, and we here briefly summarize their preparation. In general, the process of CAR T-cell manufacturing and delivery involves the following major steps (Fig. 1): (1) Isolation: Peripheral blood mononuclear cells are harvested from the patient or donor’s peripheral blood using a standard leukapheresis procedure, a process whereby blood is removed from an individual’s antecubital veins, separated into select components, and the remainder of the blood returned to the individual’s circulation [38]. (2) Modification: T cells were activated with CD3/CD28 magnetic beads (Dynabead) to be susceptible to viral transduction [39]. Then, CARs with the high affinity to predefined tumor antigens are transduced into these T cells by way of viral (lentiviral or retroviral) or nonviral (transposon) gene transfer systems. Lentiviral vectors and gammaretroviral vectors are currently two standard methods of viral transduction to equip T cells with a CAR [3840]. The nonviral transduction methods usually used in engineering CAR-T cell are plasmid DNA [41] and RNA electroporation, which are also applied to T cells without pre-activation [42]. In this step, the CARs identifying tumor-associated antigens (TAAs) and, simultaneously, activating T cells were genetically expressed on the collected T cells. (3) Expansion: The CAR-T cells are expanded ex vivo to achieve the desired modified T cell dose. (4) Reinfusion: The modified T cells amplified to clinically relevant cell numbers were subsequently reinfused to the beforehand lymphocyte-depleted patient. Then, a novel CliniMACS Prodigy (Miltenyi Biotec), an automated manufacturing of CAR-T cells, has been adapted for lentiviral transduction of T cells which exhibited enormous potential [43].

Therapeutic effect of CAR-T in different systems

Clinical trials to date have almost all focused on second- or third-generation CAR constructs. We here concluded the clinical applications of second- or third-generation CAR-T cells in different system tumors and summarized them in Table 2.
Table 2
Clinical trials of CAR-T therapy on different tumors
Tumors
scFv
Single domain
Dose (cells /kg or cells/ m2)
Clinical trials (phage and NCT number) (www.​clinicaltrials.​gov/)
Number of treated patients
Responses
Persistence
References
ALL
CD19
CD28 + CD3ζ
1.5 × 106 to 3 × 106
Phase I (NCT01044069)
5
5 CR
Uncertaina
[87]
ALL
CD19
CD137+ CD3ζ
1.4 × 106 to 1.2 × 107
Phase I (NCT01626495)
2
2 CR
One persisted 11 months, the other relapsed
[45]
ALL
CD19
41BB + CD3ζ
0.76 × 106 to 20.6 × 106
Phase I/ ΙΙ
(NCT01626495) ( NCT01029366)
30
27 CR
2 to 3 months
[48]
ALL
CD19
CD28 + CD3ζ
3 × 106
Phase I (NCT01044069)
16
14 CR
2 to 3 months
[52]
ALL
CD19
CD28 + CD3ζ
1 × 106 (maximum)
Phase I (NCT01593696)
21
12 CR
Un stated
[13]
CLL
CD19
CD137+ CD3ζ
1.5 × 105
Phase I (NCT01029366)
3
3CR
10 months
[46]
CLL
CD19
CD28 + CD3ζ
0.2–1.1 × 107
Phase I (NCT00466531)
8
1 PR
uncertain
[91]
CLL
CD19
CD28 + CD3ζ
1 × 106, 1.5 × 106, 4 × 106
Phase I (NCT01087294).
10
3 CR
< 1 month
[92]
CLL
CD19
41BB + CD3ζ
0.14 × 108 to 11 × 108
Phase I (NCT01029366)
14
4 CR, 4 PR
14 to 49 months
[49]
CLL
CD19
41BB + CD3ζ
1.6 × 107; 1.0 × 107; 1.46 × 105
Phase I (NCT01029366)
3
2 CR, 1 PR
> 6 months
[93]
CLL/NHL/MM
κ light chain
CD28 + CD3ζ
2 × 107, 1 × 108, 2 × 108
NCT00881920
16 (9 CLL/NHL, 7 MM)
2 CR, 1 PR
6 weeks
[94]
CLL
CD19
CD28 + 41BB+ CD3ζ
2 × 105, 2 × 106, or 2 × 107
unstated
24
4 CR, 10 PR
6 months
[95]
MM
CD19
CD137+ CD3ζ
1 × 107 to 5 × 107
Phase I (NCT02135406)
10
Uncertain
[54]
Lymphomas
CD19
41BB+ CD3ζ
3.08 × 106 to 8.87 × 106
NCT02030834
28
16 CR
29.3 months
[47]
NSCLC
EGFR
CD137+ CD3ζ
0.45 to 1.09 × 107
Phase I (NCT01869166)
11
2 PR, 5 SD
2 to 8 months
[20]
CCA
EGFR
CD137+ CD3ζ
2.2/2.1 × 106, 1.22 × 106
Phase I (NCT01869166) (NCT02541370)
1
1 PR
13 months
[63]
CRC
CEA
CD28/CD137+ CD3ζ, CD28+ D137+ CD3ζ
1 × 105 to 1 × 108
Phase I (NCT02349724)
10
7 SD
[33]
SVC
MUC1
CD28+  4-1BB+ CD3ζ
5 × 105
Phase I/II (NCT02587689)
1
Tumor necrosis
Unstated
[68]
GBM
GD2
unstated
2 × 107, 5 × 107, 1×  108
Phase I (NCT00085930)
19
3CR
>  6 weeks
[69]
GBM
EGFRvIII
41BB + CD3ζ
1 × 107
Phase I (NCT02209376)
10
1SD
[70]
GBM
HER2
CD28+ CD3ζ
1 × 106 to 1 × 108
Phase I (NCT01109095)
17
1 PR,
>  9 months
[71]
7 SD
GBM
IL13Ra2
41BB + CD3ζ
2 × 106, 10 × 106
Phase I (NCT02208362)
1
Tumor necrosis
7.5 months
[72]
Sarcoma
HER2
CD28+ CD3ζ
1 × 104 to 1 × 108
Phase I/II (NCT00902044)
19
4 SD
[74]
aFour of these patients were treated with subsequent HSCT

CAR-T for hematological malignancy treatment

CAR-T cell therapy is perhaps best known for its role in the treatment of B-cell hematologic malignancies. CD19, a surface protein highly expressed on most B-lineage lymphocytes and not on normal tissue outside the B-lineage [44], is the most thoroughly studied target in all of the hematological malignancy-associated antigens, and CD19-specific CAR-T cell therapy has demonstrated enormous efficiency in inducing endurable remissions of several hematological malignancies, including ALL, chronic lymphocytic leukemia (CLL), and NHL [4547], with complete remissions (CR) in ALL at 90% and response rates in CLL greater than 50% [48, 49]. In 2008, Till et al. reported that CD20-targeted CAR-T cells have demonstrated potential antitumor activity in treating indolent NHL and mantle cell lymphoma [50]. Later, in 2010, Kochenderfer et al. treated a patient with advanced follicular lymphoma with anti-CD19-CAR-transduced T cells, and the patient underwent a dramatic regression [51]. In 2011, Porter et al. designed a second-generation CAR-T cell in treating a patient with refractory CLL, and all three underwent CR. These findings provoked research exploring the antitumor efficacy of CD19-redirected T cells for B-cell neoplasms [46]. Based on these, in 2013, Grupp et al. extended the application of CAR-T cells to refractory B-cell ALL and established a clinical trial involving two children with ALL treated with CTL019 CAR-T cells. Surprisingly, CR was observed in both patients, demonstrating that CAR-T cells may be favorable for the treatment of patients with refractory ALL. However, cytokine-release syndrome (CRS) was also observed [45]. Subsequently, in 2014, Maude et al. conducted pilot clinical trials of 30 patients (children and adults) with r/r CD19+ ALL, in which infused autologous T cells are transduced with a CTL019. CR was achieved in 27 patients. CTL019 was effective in treating r/r ALL, even in stem cell transplantation-failed patients. Nevertheless, CRS was developed in all the patients [48]. Furthermore, Davila et al. treated 16 adult patients with r/r ALL with 19-28z CAR-T cells specific to the CD19 antigen and achieved a promising outcome, with overall CR rate of 88% (14/16). CRS, which may be related to a systemic inflammatory process induced by the reaction between infused CAR T cells and the targeted CD19 antigen, is almost inevitable. Therefore, the diagnostic criteria for severe CRS were defined, and serum C-reactive protein can serve as a reliable indicator for CRS severity [52]. Although CD19 is not an ideal antigen in multiple myeloma (MM), for its low expression in MM [53], Garfall et al. still reported that CAR-T cell therapy in conjunction with autologous transplantation has achieved durable CR in a patient with advanced MM [54]. These findings provided a road map for application of CAR-T cells in solid tumors.

CAR-T for solid tumor treatment

The unprecedented success of CAR-T cell therapy in hematological malignancy fostered the enthusiasm to expand this technology to solid tumors. However, this therapy encountered some difficulties in application for solid malignancy. The reasons for this phenomenon are as follows: (1) lack of eligible, effective targets such as CD19 because most target antigens are more or less expressed in normal tissues; (2) hostile immunosuppressive microenvironment of solid tumors that affect the T cells; and (3) heterogeneity of solid tumors. Although the exploration of CAR-T cell treatment in solid tumors is not as definitive as in the research of hematological malignancy, some studies have achieved promising outcomes. Here, we introduced some diseases in which the CAR-T-cell treatment has exhibited benign clinical responses.

NSCLC

Advanced strategies, including surgery, radiotherapy, chemotherapy, and targeted therapy, have improved the survival in patients with NSCLC. Nevertheless, the 5-year survival rate of late-stage NSCLC is still unsatisfactory [55]. The breakthrough treatment of immunotherapy with CAR-T cells in hematology raised the possibility of their use in NSCLC. In 2016, Feng et al. first studied the safety and feasibility of epidermal growth factor receptor (EGFR)-targeted CAR-T cell therapy in treating 11 patients with advanced r/r NSCLC. Two patients obtained partial response (PR), and five had stable diseases (SD) after the infusion of CART-EGFR cells with mild side effects--mild skin toxicity, nausea, vomiting, dyspnea and hypotension [20]. In addition, other TAAs, like erythropoietin-producing hepatocellular carcinoma A2 (EphA2) (Li et al. 2017) [56], prostate stem cell antigen (PSCA), and mucin 1 (MUC1) (Wei et al. 2017) [57], have also been detected in NSCLC and confirmed to be promising targeting antigen for CAR-T cells. These antigen-targeted CAR-T cells have been observed to cause tumor cell lysis in vitro exerting antitumor activity in xenograft mouse models. Furthermore, targeting the combination of PSCA and MUC1 can further enhance the antitumor efficacy of CAR T cells [57].

MPM

MPM is an aggressive malignancy with a median survival of less than one year [58]. Mesothelin plays an important role in screening and detecting the progression of MPM [59]. Basing on this finding, some researchers considered that CAR-T targeting mesothelin can treat MPM. A phase I clinical trial conducted at the University of Pennsylvania was designed to evaluate the manufacturing feasibility and safety of mRNA-transduced CAR T cells that target mesothelin (CART-meso cells) in patients with advanced MPM. In this study, CART-meso cells showed potent antitumor activity with no distinct on target/off-tumor toxicities (pleuritis, pericarditis, or peritonitis) [60].

Digestive system neoplasm

Cholangiocarcinoma (CCA) is a relatively rare and aggressive malignancy of the biliary tract and is characterized by late diagnosis and poor outcomes [61]. Complete surgical resection can be used as treatment. However, most of the patients will eventually relapse because of the delayed diagnosis and advanced stage of the disease [62]. In 2017, Feng et al. applied EGFR- and CD133-specific CAR-T sequential treatments as CAR-T cocktail immunotherapy for patients with advanced unresectable/metastatic CCA. An 8.5-month PR from the initial CAR-T-EGFR treatment and another 4.5-month PR from the subsequent CD133-specific CAR-T immunotherapy were obtained. However, the epidermal and endothelial damages caused by the infusion of CAR-T cells cannot be disregarded, thereby requiring further investigation [63].
A phase I clinical trial conducted at the University of Pennsylvania was designed to evaluate the manufacturing feasibility and safety of CART-meso cells in patients with advanced MPM and explore the antitumor effect of CART-meso cells in patients with pancreatic cancer. The results showed the antitumor activity. CART-meso cells were also detected in primary and metastatic tumor sites by collecting ascites and conducting a tumor biopsy [60].
Zhang et al. established a clinical trial of CEA CAR-T therapy of 10 patients with CRC by systemic delivery through intravenous (IV) infusion to evaluate its safety and efficacy. Out of the 10 patients, 7 patients who experienced progressive disease in the previous treatments have SD after the CAR-T therapy. Moreover, severe adverse events related to CAR-T therapy are not observed [33].

Genitourinary system diseases

Epithelial ovarian cancer (EOC) remains to be the most mortal of all gynecological malignancies mainly due to its subtle nature. Despite the fact that most patients with EOC yield a good clinical response following current advanced therapy, almost all patients will ultimately relapse and eventually develop drug resistance [64]. The survival of patients with EOC is positively related to the presence of TILs, which play a significant role in adoptive T-cell therapy [65]. MUC16, a well-known ovarian tumor antigen, is overexpressed by a majority of EOC but at a low level on normal tissues [66]. On the basis of this rationale, Brentjens et al. developed T cells expressing MUC16 to treat EOC. Moreover, to overcome the hostile tumor environment, they co-expressed IL-12 on T cells. Hence, a clinical trial testing the safety of IV and intraperitoneal infusion of genetically modified autologous T cells expressing MUC16 and secreting IL-12 in patients with EOC was conducted. The result demonstrated that the intraperitoneal injections of CAR T cells are superior to that of IV alone [67].
You et al. launched a phase I clinical trial to evaluate the ability of engineered CAR-T cells targeting MUC1 to treat patients with seminal vesicle cancer (SVC). To suppress the unfavorable tumor microenvironment, they induced IL-12 co-expression and constructed two anti-MUC1 CAR-T cell lines, that is, SM3-CAR (co-expressing IL-12) and pSM3-CAR (without IL-12). These two types of CAR-T cells were injected intratumorally into two separate metastatic lesions of the same patient with MUC1+ SVC as part of an interventional treatment strategy. The results showed tumor necrosis induced by pSM3-CAR is more evident than that by SM3-CAR, without significant side effects [68].

GBM

CAR-T cell has also been explored in recent years to treat central nervous system cancers. In 2011, Louis et al. conducted a clinical trial of GD2-specific CAR-T therapy in 19 patients with high-risk neuroblastoma. Three patients had a CR to CAR-T cell infusion, with only slight fever and light-to-moderate local pain being observed [69]. In 2017, a clinical trial of IV administration of EGFRvIII-specific CAR-T cells for the treatment of 10 patients with refractory GBM was established at the University of Pennsylvania. The infusion of CAR-T cells was feasible and safe, without evident off-tumor toxicity or CRS [70]. Furthermore, an open-labeled phase 1 dose-escalation study was conducted at the Baylor College of Medicine, Houston Methodist Hospital, and Texas Children’s Hospital to evaluate the safety and anti-GBM activity of HER2-specific CAR-modified virus-specific T cells in patients with progressive GBM. The results showed that the infusions are well tolerated, with no dose-limiting toxic effects. Moreover, 1 patient showed a PR for more than 9 months, whereas 7 patients had SD for 8 weeks to 29 months [71]. In addition, Brown et al. initiated a clinical trial with one patient with relapsing GBM, who received administration of IL13Rα2 targeted IL13BBζ–CAR-T cells, and regression of tumors was observed and persisted for 7.5 months after the administration of CAR-T cell therapy [72].

Sarcomas

Sarcoma, which can be located anywhere in the body, is usually treated with surgical resection, with or without radiotherapy, and chemotherapy. However, patients with advanced stage sarcomas still have poor prognosis [73]. Hence, several researchers speculated that CAR-T-cell treatment may benefit patients with sarcoma. In 2015, Ahmed et al. designed a phase I/ II clinical study to evaluate the safety and efficacy of HER2-specific CAR-T cells in patients with r/r HER2-positive sarcoma. A total of 19 patients were enrolled in this research, and they received escalating doses of HER2-specific CAR-T cells. Although no CR were observed, 4 out of 17 patients that can be evaluated have SD for 12 weeks to 14 months [74].

Conclusion

In this review, we summarized the current clinical studies on CAR-T treatment of hematologic diseases and solid tumors. Clinical outcomes of CAR-T cell therapy in patients with hematologic malignancies have been encouraging. However, in patients with solid tumors, the outcomes have been discouraging, nevertheless, not gloomy. CAR-T cell therapy, as a promising treatment, has the following advantages: (1) binding surface antigen of tumors in non-MHC restriction manner; (2) recognizing multiple antigens simultaneously; and (3) obtaining a large number of CAR-T cells ex vivo in a short term. CRS is an ineluctable complication of CAR-T-cell therapy on the basis of the clinical trials of hematological malignancies mentioned above. The manifestations of CRS include fevers, hypotension, nausea, myalgias, and neurologic dysfunction. When CRS is severe, vasopressors, mechanical ventilation, antiepileptics, and hemodialysis may be required [52]. Fortunately, researchers can now control most cases of CRS with an anti-interleukin 6 antibody, such as tocilizumab, which was approved by the FDA for the treatment of CAR-T-cell therapy-induced CRS in August 2017 [75]. The CRS is not common in solid tumors treated by CAR-T cells; however, on target/off-tumor toxicity has become common due to unavoidable expression, to some extent, of target antigens in normal tissues [60]. This phenomenon could be solved by manufacturing CAR-T cells with dual antigen specificity or switchable dual-receptor [76, 77] or by transfecting T cells with mRNA encoding CAR to reduce their half-life; these CAR-T cells can be repeated administered, and the toxicity to normal tissues can be mitigated [78]. Moreover, the hostile immunosuppressive microenvironment is one of the major challenges in CAR-T-cell treatment of solid tumors. The tumor microenvironment is a complex and dense fibrotic matrix network composed of malignant and nonmalignant cells, in which the infiltrated CAR-T cells can be inhibited by immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressive cells [79]. In a recent study conducted by Chen et al., CARs were engineered to target a bunch of soluble ligands, including TGF-β, an otherwise immunosuppressive factor in a variety of solid tumors, and demonstrated the ability to effectively convert TGF-β from a potent immunosuppressive cytokine to a strong stimulant for the primary human T cells [80]. Another research by Batchu et al. also shed light on the immunosuppressive microenvironment in solid tumors. They discovered that suppressing interleukin-10, an immune inhibitory cytokine secreted by Tregs and pancreatic cancer cells, can reverse the negative effect of the tumor microenvironment on mesothelin-CAR-T cells in pancreatic cancer in vitro [81]. In addition, solid tumors have relatively limited body distribution and are concrete compared with hematological malignancies. Hence, we can hypothesize that in some cases, the regional delivery of CAR-T cells may be immensely superior to systemic administration. Several studies reported that local injection, such as intrapleural administration and local intracranial delivery, show greater potential than IV injection [23, 82]. These findings indicated that CAR-T-cell therapy can gain momentum to break through the restriction of tumor microenvironment in treating solid malignancies. In the aggregates, CAR-T-cell therapy is a promising strategy against neoplasms. Several key points must be considered to translate the success of CAR-T cell therapy to extensive solid tumors. These widely acknowledged key points include finding a specific antigen or engineering multiple antigen-targeted CAR-T cells, directly targeting the constituent of immunosuppressive microenvironment, and creating a suitable tumor microenvironment [83]. In addition to these factors, replacing IV infusion with regional delivery, transfecting T cells with mRNA encoding CAR, and combining T cells with oncolytic viruses or immune-checkpoint blockade to bolster the potency of CAR-T cells can also be considered.

Acknowledgements

This work was supported by the National Natural Science Foundation of China [No.81572976], the China Postdoctoral Science Foundation [Nos.2016M590505, 2017T100407], the Jiangsu Provincial Medical Talent Foundation, the Postgraduate Research & Practice Innovation Program of Jiangsu Province [No. KYCX17_1715, No. KYCX17_1718].
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Ruhlmann C, Iversen T, Okera M, Muhic A, Kristensen G, Feyer P, Hansen O, Herrstedt J. Multinational study exploring patients’ perceptions of side-effects induced by chemo-radiotherapy. Radiother Oncol. 2015;117:333–7.CrossRefPubMed Ruhlmann C, Iversen T, Okera M, Muhic A, Kristensen G, Feyer P, Hansen O, Herrstedt J. Multinational study exploring patients’ perceptions of side-effects induced by chemo-radiotherapy. Radiother Oncol. 2015;117:333–7.CrossRefPubMed
2.
Zurück zum Zitat Neuman B, Ailon T, Scheer J, Klineberg E, Sciubba D, Jain A, Zebala L, Passias P, Daniels A, Burton D, et al. Development and validation of a novel adult spinal deformity surgical invasiveness score: analysis of 464 patients. Neurosurgery. 2017. https://doi.org/10.1093/neuros/nyx303. Neuman B, Ailon T, Scheer J, Klineberg E, Sciubba D, Jain A, Zebala L, Passias P, Daniels A, Burton D, et al. Development and validation of a novel adult spinal deformity surgical invasiveness score: analysis of 464 patients. Neurosurgery. 2017. https://​doi.​org/​10.​1093/​neuros/​nyx303.
3.
Zurück zum Zitat Yin Q, Shen J, Zhang Z, Yu H, Li Y. Reversal of multidrug resistance by stimuli-responsive drug delivery systems for therapy of tumor. Adv Drug Deliv Rev. 2013;65:1699–715.CrossRefPubMed Yin Q, Shen J, Zhang Z, Yu H, Li Y. Reversal of multidrug resistance by stimuli-responsive drug delivery systems for therapy of tumor. Adv Drug Deliv Rev. 2013;65:1699–715.CrossRefPubMed
4.
Zurück zum Zitat Li R, Pu X, Chang J, Ye Y, Komaki R, Minna J, Roth J, Han B, Wu X. MiRNA-related genetic variations associated with radiotherapy-induced toxicities in patients with locally advanced non-small cell lung Cancer. PLoS One. 2016;11:e0150467.CrossRefPubMedPubMedCentral Li R, Pu X, Chang J, Ye Y, Komaki R, Minna J, Roth J, Han B, Wu X. MiRNA-related genetic variations associated with radiotherapy-induced toxicities in patients with locally advanced non-small cell lung Cancer. PLoS One. 2016;11:e0150467.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Crompton J, Klemen N, Kammula U. Metastasectomy for tumor-infiltrating lymphocytes: an emerging operative indication in surgical oncology. Ann Surg Oncol. 2017; Crompton J, Klemen N, Kammula U. Metastasectomy for tumor-infiltrating lymphocytes: an emerging operative indication in surgical oncology. Ann Surg Oncol. 2017;
6.
Zurück zum Zitat Kunert A, Obenaus M, Lamers C, Blankenstein T, Debets R. T-cell receptors for clinical therapy: in vitro assessment of toxicity risk. Clin Cancer Res. 2017;23:6012–20.CrossRefPubMed Kunert A, Obenaus M, Lamers C, Blankenstein T, Debets R. T-cell receptors for clinical therapy: in vitro assessment of toxicity risk. Clin Cancer Res. 2017;23:6012–20.CrossRefPubMed
7.
Zurück zum Zitat Hay K, Turtle C. Chimeric antigen receptor (CAR) T cells: lessons learned from targeting of CD19 in B-cell malignancies. Drugs. 2017;77:237–45.CrossRefPubMedPubMedCentral Hay K, Turtle C. Chimeric antigen receptor (CAR) T cells: lessons learned from targeting of CD19 in B-cell malignancies. Drugs. 2017;77:237–45.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Wu R, Forget M, Chacon J, Bernatchez C, Haymaker C, Chen J, Hwu P, Radvanyi L. Adoptive T-cell therapy using autologous tumor-infiltrating lymphocytes for metastatic melanoma: current status and future outlook. Cancer J. 2012;18:160–75.CrossRefPubMedPubMedCentral Wu R, Forget M, Chacon J, Bernatchez C, Haymaker C, Chen J, Hwu P, Radvanyi L. Adoptive T-cell therapy using autologous tumor-infiltrating lymphocytes for metastatic melanoma: current status and future outlook. Cancer J. 2012;18:160–75.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Feldman S, Assadipour Y, Kriley I, Goff S, Rosenberg S. Adoptive cell therapy--tumor-infiltrating lymphocytes, T-cell receptors, and chimeric antigen receptors. Semin Oncol. 2015;42:626–39.CrossRefPubMed Feldman S, Assadipour Y, Kriley I, Goff S, Rosenberg S. Adoptive cell therapy--tumor-infiltrating lymphocytes, T-cell receptors, and chimeric antigen receptors. Semin Oncol. 2015;42:626–39.CrossRefPubMed
10.
Zurück zum Zitat Mazza C, Malissen B. What guides MHC-restricted TCR recognition? Semin Immunol. 2007;19:225–35.CrossRefPubMed Mazza C, Malissen B. What guides MHC-restricted TCR recognition? Semin Immunol. 2007;19:225–35.CrossRefPubMed
11.
Zurück zum Zitat Harris D, Kranz D, Cell Therapies AT, Comparison A. Of T cell receptors and chimeric antigen receptors. Trends Pharmacol Sci. 2016;37:220–30.CrossRefPubMed Harris D, Kranz D, Cell Therapies AT, Comparison A. Of T cell receptors and chimeric antigen receptors. Trends Pharmacol Sci. 2016;37:220–30.CrossRefPubMed
12.
Zurück zum Zitat Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86:10024–8.CrossRefPubMedPubMedCentral Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86:10024–8.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385:517–28.CrossRefPubMed Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385:517–28.CrossRefPubMed
14.
Zurück zum Zitat Ghorashian S, Pule M, Amrolia P. CD19 chimeric antigen receptor T cell therapy for haematological malignancies. Br J Haematol. 2015;169:463–78.CrossRefPubMed Ghorashian S, Pule M, Amrolia P. CD19 chimeric antigen receptor T cell therapy for haematological malignancies. Br J Haematol. 2015;169:463–78.CrossRefPubMed
15.
Zurück zum Zitat Park J, Geyer M, Brentjens R. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016;127:3312–20.CrossRefPubMedPubMedCentral Park J, Geyer M, Brentjens R. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016;127:3312–20.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Morello A, Sadelain M, Adusumilli P. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6:133–46.CrossRefPubMed Morello A, Sadelain M, Adusumilli P. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6:133–46.CrossRefPubMed
17.
Zurück zum Zitat Gill S, June C. Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol Rev. 2015;263:68–89.CrossRefPubMed Gill S, June C. Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol Rev. 2015;263:68–89.CrossRefPubMed
18.
Zurück zum Zitat Ruella M, Kenderian S. Next-generation chimeric antigen receptor T-cell therapy: going off the shelf. BioDrugs. 2017;31:473–81.CrossRefPubMed Ruella M, Kenderian S. Next-generation chimeric antigen receptor T-cell therapy: going off the shelf. BioDrugs. 2017;31:473–81.CrossRefPubMed
19.
Zurück zum Zitat Neelapu S, Locke F, Bartlett N, Lekakis L, Miklos D, Jacobson C, Braunschweig I, Oluwole O, Siddiqi T, Lin Y, et al. Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–44.CrossRefPubMedPubMedCentral Neelapu S, Locke F, Bartlett N, Lekakis L, Miklos D, Jacobson C, Braunschweig I, Oluwole O, Siddiqi T, Lin Y, et al. Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–44.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Feng K, Guo Y, Dai H, Wang Y, Li X, Jia H, Han W. Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci. 2016;59:468–79.CrossRefPubMed Feng K, Guo Y, Dai H, Wang Y, Li X, Jia H, Han W. Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci. 2016;59:468–79.CrossRefPubMed
21.
Zurück zum Zitat Lamers C, Klaver Y, Gratama J, Sleijfer S, Debets R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study overview. Biochem Soc Trans. 2016;44:951–9.CrossRefPubMed Lamers C, Klaver Y, Gratama J, Sleijfer S, Debets R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study overview. Biochem Soc Trans. 2016;44:951–9.CrossRefPubMed
22.
Zurück zum Zitat Brown CE, Aguilar B, Starr R, Yang X, Chang WC, Weng L, Chang B, Sarkissian A, Brito A, Sanchez JF, et al. Optimization of IL13Ralpha2-targeted chimeric antigen receptor T cells for improved anti-tumor efficacy against glioblastoma. Mol Ther. 2017; Brown CE, Aguilar B, Starr R, Yang X, Chang WC, Weng L, Chang B, Sarkissian A, Brito A, Sanchez JF, et al. Optimization of IL13Ralpha2-targeted chimeric antigen receptor T cells for improved anti-tumor efficacy against glioblastoma. Mol Ther. 2017;
23.
Zurück zum Zitat Adusumilli P, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, Jones D, Sadelain M. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6:261ra151.CrossRefPubMedPubMedCentral Adusumilli P, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, Jones D, Sadelain M. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6:261ra151.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Letourneur F, Klausner R. T-cell and basophil activation through the cytoplasmic tail of T-cell-receptor zeta family proteins. Proc Natl Acad Sci U S A. 1991;88:8905–9.CrossRefPubMedPubMedCentral Letourneur F, Klausner R. T-cell and basophil activation through the cytoplasmic tail of T-cell-receptor zeta family proteins. Proc Natl Acad Sci U S A. 1991;88:8905–9.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Eshhar Z, Waks T, Gross G, Schindler D. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90:720–4.CrossRefPubMedPubMedCentral Eshhar Z, Waks T, Gross G, Schindler D. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90:720–4.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kershaw M, Westwood J, Parker L, Wang G, Eshhar Z, Mavroukakis S, White D, Wunderlich J, Canevari S, Rogers-Freezer L, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–15.CrossRefPubMedPubMedCentral Kershaw M, Westwood J, Parker L, Wang G, Eshhar Z, Mavroukakis S, White D, Wunderlich J, Canevari S, Rogers-Freezer L, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–15.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat van der Stegen S, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov. 2015;14:499–509.CrossRefPubMed van der Stegen S, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov. 2015;14:499–509.CrossRefPubMed
28.
Zurück zum Zitat Savoldo B, Ramos C, Liu E, Mims M, Keating M, Carrum G, Kamble R, Bollard C, Gee A, Mei Z, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121:1822–6.CrossRefPubMedPubMedCentral Savoldo B, Ramos C, Liu E, Mims M, Keating M, Carrum G, Kamble R, Bollard C, Gee A, Mei Z, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121:1822–6.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Till B, Jensen M, Wang J, Qian X, Gopal A, Maloney D, Lindgren C, Lin Y, Pagel J, Budde L, 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.CrossRefPubMedPubMedCentral Till B, Jensen M, Wang J, Qian X, Gopal A, Maloney D, Lindgren C, Lin Y, Pagel J, Budde L, 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.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM, Varela-Rohena A, Haines KM, Heitjan DF, Albelda SM, et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A. 2009;106:3360–5.CrossRefPubMedPubMedCentral Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM, Varela-Rohena A, Haines KM, Heitjan DF, Albelda SM, et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A. 2009;106:3360–5.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Wang J, Jensen M, Lin Y, Sui X, Chen E, Lindgren C, Till B, Raubitschek A, Forman S, Qian X, et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther. 2007;18:712–25.CrossRefPubMed Wang J, Jensen M, Lin Y, Sui X, Chen E, Lindgren C, Till B, Raubitschek A, Forman S, Qian X, et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther. 2007;18:712–25.CrossRefPubMed
32.
Zurück zum Zitat Haso W, Lee D, Shah N, Stetler-Stevenson M, Yuan C, Pastan I, Dimitrov D, Morgan R, FitzGerald D, Barrett D, et al. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121:1165–74.CrossRefPubMedPubMedCentral Haso W, Lee D, Shah N, Stetler-Stevenson M, Yuan C, Pastan I, Dimitrov D, Morgan R, FitzGerald D, Barrett D, et al. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121:1165–74.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Zhang C, Wang Z, Yang Z, Wang M, Li S, Li Y, Zhang R, Xiong Z, Wei Z, Shen J, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA(+) metastatic colorectal cancers. Mol Ther. 2017;25:1248–58.CrossRefPubMedPubMedCentral Zhang C, Wang Z, Yang Z, Wang M, Li S, Li Y, Zhang R, Xiong Z, Wei Z, Shen J, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA(+) metastatic colorectal cancers. Mol Ther. 2017;25:1248–58.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Chmielewski M, Hombach A, Abken H. Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol Rev. 2014;257:83–90.CrossRefPubMed Chmielewski M, Hombach A, Abken H. Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol Rev. 2014;257:83–90.CrossRefPubMed
35.
Zurück zum Zitat Chmielewski M, Kopecky C, Hombach A, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011;71:5697–706.CrossRefPubMed Chmielewski M, Kopecky C, Hombach A, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011;71:5697–706.CrossRefPubMed
36.
Zurück zum Zitat Hombach A, Hombach A, Abken H. Adoptive immunotherapy with genetically engineered T cells: modification of the IgG1 Fc ‘spacer’ domain in the extracellular moiety of chimeric antigen receptors avoids ‘off-target’ activation and unintended initiation of an innate immune response. Gene Ther. 2010;17:1206–13.CrossRefPubMed Hombach A, Hombach A, Abken H. Adoptive immunotherapy with genetically engineered T cells: modification of the IgG1 Fc ‘spacer’ domain in the extracellular moiety of chimeric antigen receptors avoids ‘off-target’ activation and unintended initiation of an innate immune response. Gene Ther. 2010;17:1206–13.CrossRefPubMed
37.
Zurück zum Zitat D'Aloia M, Caratelli S, Palumbo C, Battella S, Arriga R, Lauro D, Palmieri G, Sconocchia G, Alimandi M. T lymphocytes engineered to express a CD16-chimeric antigen receptor redirect T-cell immune responses against immunoglobulin G-opsonized target cells. Cytotherapy. 2016;18:278–90.CrossRefPubMed D'Aloia M, Caratelli S, Palumbo C, Battella S, Arriga R, Lauro D, Palmieri G, Sconocchia G, Alimandi M. T lymphocytes engineered to express a CD16-chimeric antigen receptor redirect T-cell immune responses against immunoglobulin G-opsonized target cells. Cytotherapy. 2016;18:278–90.CrossRefPubMed
38.
Zurück zum Zitat McGuirk J, Waller EK, Qayed M, Abhyankar S, Ericson S, Holman P, Keir C, Myers GD. Building blocks for institutional preparation of CTL019 delivery. Cytotherapy. 2017;19:1015–24.CrossRefPubMed McGuirk J, Waller EK, Qayed M, Abhyankar S, Ericson S, Holman P, Keir C, Myers GD. Building blocks for institutional preparation of CTL019 delivery. Cytotherapy. 2017;19:1015–24.CrossRefPubMed
39.
Zurück zum Zitat Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C, Yeh R, Capacio V, Olszewska M, Hosey J, et al. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009;32:169–80.CrossRefPubMedPubMedCentral Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C, Yeh R, Capacio V, Olszewska M, Hosey J, et al. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009;32:169–80.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B, Danet-Desnoyers G, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17:1453–64.CrossRefPubMedPubMedCentral Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B, Danet-Desnoyers G, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17:1453–64.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Schmeer M, Buchholz T, Schleef M. Plasmid DNA manufacturing for indirect and direct clinical applications. Hum Gene Ther. 2017;28:856–61.CrossRefPubMed Schmeer M, Buchholz T, Schleef M. Plasmid DNA manufacturing for indirect and direct clinical applications. Hum Gene Ther. 2017;28:856–61.CrossRefPubMed
42.
Zurück zum Zitat Riet T, Holzinger A, Dörrie J, Schaft N, Schuler G, Abken H, Nonviral RNA. Transfection to transiently modify T cells with chimeric antigen receptors for adoptive therapy. Methods Mol Biol. 2013;969:187–201.CrossRefPubMed Riet T, Holzinger A, Dörrie J, Schaft N, Schuler G, Abken H, Nonviral RNA. Transfection to transiently modify T cells with chimeric antigen receptors for adoptive therapy. Methods Mol Biol. 2013;969:187–201.CrossRefPubMed
43.
Zurück zum Zitat Mock U, Nickolay L, Philip B, Cheung GW, Zhan H, Johnston IC, Kaiser AD, Peggs K, Pule M, Thrasher AJ, Qasim W. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy. 2016;18:1002–11.CrossRefPubMed Mock U, Nickolay L, Philip B, Cheung GW, Zhan H, Johnston IC, Kaiser AD, Peggs K, Pule M, Thrasher AJ, Qasim W. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy. 2016;18:1002–11.CrossRefPubMed
44.
Zurück zum Zitat Scheuermann R, Racila E. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk Lymphoma. 1995;18:385–97.CrossRefPubMed Scheuermann R, Racila E. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk Lymphoma. 1995;18:385–97.CrossRefPubMed
45.
Zurück zum Zitat Grupp S, Kalos M, Barrett D, Aplenc R, Porter D, Rheingold S, Teachey D, Chew A, Hauck B, Wright J, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–18.CrossRefPubMedPubMedCentral Grupp S, Kalos M, Barrett D, Aplenc R, Porter D, Rheingold S, Teachey D, Chew A, Hauck B, Wright J, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–18.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Porter D, Levine B, Kalos M, Bagg A, June C. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–33.CrossRefPubMedPubMedCentral Porter D, Levine B, Kalos M, Bagg A, June C. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–33.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak O, Brogdon JL, Pruteanu-Malinici I, Bhoj V, Landsburg D, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017; Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak O, Brogdon JL, Pruteanu-Malinici I, Bhoj V, Landsburg D, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;
48.
Zurück zum Zitat Maude S, Frey N, Shaw P, Aplenc R, Barrett D, Bunin N, Chew A, Gonzalez V, Zheng Z, Lacey S, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–17.CrossRefPubMedPubMedCentral Maude S, Frey N, Shaw P, Aplenc R, Barrett D, Bunin N, Chew A, Gonzalez V, Zheng Z, Lacey S, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–17.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Porter D, Hwang W, Frey N, Lacey S, Shaw P, Loren A, Bagg A, Marcucci K, Shen A, Gonzalez V, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7:303ra139.CrossRefPubMedPubMedCentral Porter D, Hwang W, Frey N, Lacey S, Shaw P, Loren A, Bagg A, Marcucci K, Shen A, Gonzalez V, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7:303ra139.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Till B, Jensen M, Wang J, Chen E, Wood B, Greisman H, Qian X, James S, Raubitschek A, Forman S, et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood. 2008;112:2261–71.CrossRefPubMedPubMedCentral Till B, Jensen M, Wang J, Chen E, Wood B, Greisman H, Qian X, James S, Raubitschek A, Forman S, et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood. 2008;112:2261–71.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I, Raffeld M, Nathan DA, Lanier BJ, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116:4099–102.CrossRefPubMedPubMedCentral Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I, Raffeld M, Nathan DA, Lanier BJ, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116:4099–102.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O, Olszewska M, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra225.CrossRef Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O, Olszewska M, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra225.CrossRef
53.
Zurück zum Zitat Tembhare P, Yuan C, Venzon D, Braylan R, Korde N, Manasanch E, Zuchlinsky D, Calvo K, Kurlander R, Bhutani M, et al. Flow cytometric differentiation of abnormal and normal plasma cells in the bone marrow in patients with multiple myeloma and its precursor diseases. Leuk Res. 2014;38:371–6.CrossRefPubMed Tembhare P, Yuan C, Venzon D, Braylan R, Korde N, Manasanch E, Zuchlinsky D, Calvo K, Kurlander R, Bhutani M, et al. Flow cytometric differentiation of abnormal and normal plasma cells in the bone marrow in patients with multiple myeloma and its precursor diseases. Leuk Res. 2014;38:371–6.CrossRefPubMed
54.
Zurück zum Zitat Garfall A, Maus M, Hwang W, Lacey S, Mahnke Y, Melenhorst J, Zheng Z, Vogl D, Cohen A, Weiss B, et al. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N Engl J Med. 2015;373:1040–7.CrossRefPubMedPubMedCentral Garfall A, Maus M, Hwang W, Lacey S, Mahnke Y, Melenhorst J, Zheng Z, Vogl D, Cohen A, Weiss B, et al. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N Engl J Med. 2015;373:1040–7.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Schild SE, Hillman SL, Tan AD, Ross HJ, McGinnis WL, Garces YA, Graham DL, Adjei AA, Jett JR. Long-term results of a trial of concurrent chemotherapy and escalating doses of radiation for Unresectable non-small cell lung Cancer: NCCTG N0028 (alliance). J Thorac Oncol. 2017;12:697–703.CrossRefPubMedPubMedCentral Schild SE, Hillman SL, Tan AD, Ross HJ, McGinnis WL, Garces YA, Graham DL, Adjei AA, Jett JR. Long-term results of a trial of concurrent chemotherapy and escalating doses of radiation for Unresectable non-small cell lung Cancer: NCCTG N0028 (alliance). J Thorac Oncol. 2017;12:697–703.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Li N, Liu S, Sun M, Chen W, Xu X, Zeng Z, Tang Y, Dong Y, Chang A, Zhao Q. Chimeric antigen receptor-modified T cells redirected to EphA2 for the immunotherapy of non-small cell lung Cancer. Transl Oncol. 2017;11:11–7.CrossRefPubMedPubMedCentral Li N, Liu S, Sun M, Chen W, Xu X, Zeng Z, Tang Y, Dong Y, Chang A, Zhao Q. Chimeric antigen receptor-modified T cells redirected to EphA2 for the immunotherapy of non-small cell lung Cancer. Transl Oncol. 2017;11:11–7.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Wei X, Lai Y, Li J, Qin L, Xu Y, Zhao R, Li B, Lin S, Wang S, Wu Q, et al. PSCA and MUC1 in non-small-cell lung cancer as targets of chimeric antigen receptor T cells. Oncoimmunology. 2017;6:e1284722.CrossRefPubMedPubMedCentral Wei X, Lai Y, Li J, Qin L, Xu Y, Zhao R, Li B, Lin S, Wang S, Wu Q, et al. PSCA and MUC1 in non-small-cell lung cancer as targets of chimeric antigen receptor T cells. Oncoimmunology. 2017;6:e1284722.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Barone E, Gemignani F, Landi S. Overexpressed genes in malignant pleural mesothelioma: implications in clinical management. J Thorac Dis. 2018;10:S369–82.CrossRefPubMedPubMedCentral Barone E, Gemignani F, Landi S. Overexpressed genes in malignant pleural mesothelioma: implications in clinical management. J Thorac Dis. 2018;10:S369–82.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat de Fonseka D, Arnold DT, Stadon L, Morley A, Keenan E, Darby M, Armstrong L, Virgo P, Maskell NA: A prospective study to investigate the role of serial serum mesothelin in monitoring mesothelioma. BMC Cancer 2018, 18:199. de Fonseka D, Arnold DT, Stadon L, Morley A, Keenan E, Darby M, Armstrong L, Virgo P, Maskell NA: A prospective study to investigate the role of serial serum mesothelin in monitoring mesothelioma. BMC Cancer 2018, 18:199.
60.
Zurück zum Zitat Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, Chew A, Zhao Y, Levine BL, Albelda SM, et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res. 2014;2:112–20.CrossRefPubMed Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, Chew A, Zhao Y, Levine BL, Albelda SM, et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res. 2014;2:112–20.CrossRefPubMed
63.
Zurück zum Zitat Feng KC, Guo YL, Liu Y, Dai HR, Wang Y, Lv HY, Huang JH, Yang QM, Han WD. Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J Hematol Oncol. 2017;10:4.CrossRefPubMedPubMedCentral Feng KC, Guo YL, Liu Y, Dai HR, Wang Y, Lv HY, Huang JH, Yang QM, Han WD. Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J Hematol Oncol. 2017;10:4.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Padmakumar S, Parayath N, Leslie F, Nair SV, Menon D, Amiji MM. Intraperitoneal chemotherapy for ovarian cancer using sustained-release implantable devices. Expert Opin Drug Deliv. 2018:1–14. Padmakumar S, Parayath N, Leslie F, Nair SV, Menon D, Amiji MM. Intraperitoneal chemotherapy for ovarian cancer using sustained-release implantable devices. Expert Opin Drug Deliv. 2018:1–14.
65.
Zurück zum Zitat Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H, Boezen HM, van der Zee AG, Daemen T, Nijman HW. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother. 2009;58:449–59.CrossRefPubMed Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H, Boezen HM, van der Zee AG, Daemen T, Nijman HW. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother. 2009;58:449–59.CrossRefPubMed
66.
Zurück zum Zitat Cheon DJ, Wang Y, Deng JM, Lu Z, Xiao L, Chen CM, Bast RC, Behringer RR. CA125/MUC16 is dispensable for mouse development and reproduction. PLoS One. 2009;4:e4675.CrossRefPubMedPubMedCentral Cheon DJ, Wang Y, Deng JM, Lu Z, Xiao L, Chen CM, Bast RC, Behringer RR. CA125/MUC16 is dispensable for mouse development and reproduction. PLoS One. 2009;4:e4675.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Koneru M, O'Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Transl Med. 2015;13:102.CrossRefPubMedPubMedCentral Koneru M, O'Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Transl Med. 2015;13:102.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat You F, Jiang L, Zhang B, Lu Q, Zhou Q, Liao X, Wu H, Du K, Zhu Y, Meng H, et al. Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated by modified anti-MUC1 chimeric antigen receptor transduced T cells. Sci China Life Sci. 2016;59:386–97.CrossRefPubMed You F, Jiang L, Zhang B, Lu Q, Zhou Q, Liao X, Wu H, Du K, Zhu Y, Meng H, et al. Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated by modified anti-MUC1 chimeric antigen receptor transduced T cells. Sci China Life Sci. 2016;59:386–97.CrossRefPubMed
69.
Zurück zum Zitat Louis C, Savoldo B, Dotti G, Pule M, Yvon E, Myers G, Rossig C, Russell H, Diouf O, Liu E, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6.CrossRefPubMedPubMedCentral Louis C, Savoldo B, Dotti G, Pule M, Yvon E, Myers G, Rossig C, Russell H, Diouf O, Liu E, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, Martinez-Lage M, Brem S, Maloney E, Shen A, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9 O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, Martinez-Lage M, Brem S, Maloney E, Shen A, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9
71.
Zurück zum Zitat Ahmed N, Brawley V, Hegde M, Bielamowicz K, Kalra M, Landi D, Robertson C, Gray TL, Diouf O, Wakefield A, et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 2017;3:1094–101.CrossRefPubMedPubMedCentral Ahmed N, Brawley V, Hegde M, Bielamowicz K, Kalra M, Landi D, Robertson C, Gray TL, Diouf O, Wakefield A, et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 2017;3:1094–101.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Brown C, Alizadeh D, Starr R, Weng L, Wagner J, Naranjo A, Ostberg J, Blanchard M, Kilpatrick J, Simpson J, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375:2561–9.CrossRefPubMedPubMedCentral Brown C, Alizadeh D, Starr R, Weng L, Wagner J, Naranjo A, Ostberg J, Blanchard M, Kilpatrick J, Simpson J, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375:2561–9.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Linch M, Miah A, Thway K, Judson I, Benson C. Systemic treatment of soft-tissue sarcoma-gold standard and novel therapies. Nat Rev Clin Oncol. 2014;11:187–202.CrossRefPubMed Linch M, Miah A, Thway K, Judson I, Benson C. Systemic treatment of soft-tissue sarcoma-gold standard and novel therapies. Nat Rev Clin Oncol. 2014;11:187–202.CrossRefPubMed
74.
Zurück zum Zitat Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, Liu E, Dakhova O, Ashoori A, Corder A, et al. Human epidermal growth factor receptor 2 (HER2) -specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J Clin Oncol. 2015;33:1688–96.CrossRefPubMedPubMedCentral Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, Liu E, Dakhova O, Ashoori A, Corder A, et al. Human epidermal growth factor receptor 2 (HER2) -specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J Clin Oncol. 2015;33:1688–96.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA, Przepiorka D, Farrell AT, Pazdur R. FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist. 2018; Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA, Przepiorka D, Farrell AT, Pazdur R. FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist. 2018;
76.
Zurück zum Zitat Bielamowicz K, Fousek K, Byrd TT, Samaha H, Mukherjee M, Aware N, Wu MF, Orange JS, Sumazin P, Man TK, et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro-Oncology. 2018;20:506–18.CrossRefPubMed Bielamowicz K, Fousek K, Byrd TT, Samaha H, Mukherjee M, Aware N, Wu MF, Orange JS, Sumazin P, Man TK, et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro-Oncology. 2018;20:506–18.CrossRefPubMed
77.
Zurück zum Zitat Zhang E, Gu J, Xue J, Lin C, Liu C, Li M, Hao J, Setrerrahmane S, Chi X, Qi W, et al. Accurate control of dual-receptor-engineered T cell activity through a bifunctional anti-angiogenic peptide. J Hematol Oncol. 2018;11:44.CrossRefPubMedPubMedCentral Zhang E, Gu J, Xue J, Lin C, Liu C, Li M, Hao J, Setrerrahmane S, Chi X, Qi W, et al. Accurate control of dual-receptor-engineered T cell activity through a bifunctional anti-angiogenic peptide. J Hematol Oncol. 2018;11:44.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Hung CF, Xu X, Li L, Ma Y, Jin Q, Viley A, Allen C, Natarajan P, Shivakumar R, Peshwa MV, Emens LA. Development of anti-human Mesothelin-targeted chimeric antigen receptor messenger RNA-transfected peripheral blood lymphocytes for ovarian Cancer therapy. Hum Gene Ther. 2018;29(5):614–25.CrossRefPubMed Hung CF, Xu X, Li L, Ma Y, Jin Q, Viley A, Allen C, Natarajan P, Shivakumar R, Peshwa MV, Emens LA. Development of anti-human Mesothelin-targeted chimeric antigen receptor messenger RNA-transfected peripheral blood lymphocytes for ovarian Cancer therapy. Hum Gene Ther. 2018;29(5):614–25.CrossRefPubMed
79.
Zurück zum Zitat Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125:5591–6.CrossRefPubMed Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125:5591–6.CrossRefPubMed
80.
Zurück zum Zitat Chang Z, Lorenzini M, Chen X, Tran U, Bangayan N, Chen Y. Rewiring T-cell responses to soluble factors with chimeric antigen receptors. Nat Chem Biol. 2018;14:317–24.CrossRefPubMedPubMedCentral Chang Z, Lorenzini M, Chen X, Tran U, Bangayan N, Chen Y. Rewiring T-cell responses to soluble factors with chimeric antigen receptors. Nat Chem Biol. 2018;14:317–24.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Batchu RB, Gruzdyn OV, Mahmud EM, Chukr F, Dachepalli R, Manmari SK, Mostafa G, Weaver DW, Gruber SA. Inhibition of Interleukin-10 in the tumor microenvironment can restore mesothelin chimeric antigen receptor T cell activity in pancreatic cancer in vitro. Surgery. 2018;163:627–32.CrossRefPubMed Batchu RB, Gruzdyn OV, Mahmud EM, Chukr F, Dachepalli R, Manmari SK, Mostafa G, Weaver DW, Gruber SA. Inhibition of Interleukin-10 in the tumor microenvironment can restore mesothelin chimeric antigen receptor T cell activity in pancreatic cancer in vitro. Surgery. 2018;163:627–32.CrossRefPubMed
82.
Zurück zum Zitat Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK, Chang WC, Ostberg JR, Neman J, Jandial R, et al. Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2(+) breast Cancer metastasis to the brain. Clin Cancer Res. 2018;24:95–105.CrossRefPubMed Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK, Chang WC, Ostberg JR, Neman J, Jandial R, et al. Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2(+) breast Cancer metastasis to the brain. Clin Cancer Res. 2018;24:95–105.CrossRefPubMed
83.
84.
Zurück zum Zitat Hege K, Bergsland E, Fisher G, Nemunaitis J, Warren R, McArthur J, Lin A, Schlom J, June C, Sherwin S. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.CrossRefPubMedPubMedCentral Hege K, Bergsland E, Fisher G, Nemunaitis J, Warren R, McArthur J, Lin A, Schlom J, June C, Sherwin S. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat Park J, Digiusto D, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet H, Bautista C, Chang W, Ostberg J, Jensen M. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–33.CrossRefPubMed Park J, Digiusto D, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet H, Bautista C, Chang W, Ostberg J, Jensen M. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–33.CrossRefPubMed
86.
Zurück zum Zitat Kowolik C, Topp M, Gonzalez S, Pfeiffer T, Olivares S, Gonzalez N, Smith D, Forman S, Jensen M, Cooper L. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006;66:10995–1004.CrossRefPubMed Kowolik C, Topp M, Gonzalez S, Pfeiffer T, Olivares S, Gonzalez N, Smith D, Forman S, Jensen M, Cooper L. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006;66:10995–1004.CrossRefPubMed
87.
Zurück zum Zitat Brentjens R, Davila M, Riviere I, Park J, Wang X, Cowell L, Bartido S, Stefanski J, Taylor C, Olszewska M, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra138.CrossRef Brentjens R, Davila M, Riviere I, Park J, Wang X, Cowell L, Bartido S, Stefanski J, Taylor C, Olszewska M, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra138.CrossRef
88.
Zurück zum Zitat Kandalaft L, Powell D, 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.CrossRefPubMedPubMedCentral Kandalaft L, Powell D, 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.CrossRefPubMedPubMedCentral
89.
Zurück zum Zitat Tang XY, Sun Y, Zhang A, Hu GL, Cao W, Wang DH, Zhang B, Chen H. Third-generation CD28/4-1BB chimeric antigen receptor T cells for chemotherapy relapsed or refractory acute lymphoblastic leukaemia: a non-randomised, open-label phase I trial protocol. BMJ Open. 2016;6:e013904.CrossRefPubMedPubMedCentral Tang XY, Sun Y, Zhang A, Hu GL, Cao W, Wang DH, Zhang B, Chen H. Third-generation CD28/4-1BB chimeric antigen receptor T cells for chemotherapy relapsed or refractory acute lymphoblastic leukaemia: a non-randomised, open-label phase I trial protocol. BMJ Open. 2016;6:e013904.CrossRefPubMedPubMedCentral
90.
Zurück zum Zitat Zhong XS, 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. Mol Ther. 2010;18:413–20.CrossRefPubMed Zhong XS, 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. Mol Ther. 2010;18:413–20.CrossRefPubMed
91.
Zurück zum Zitat Brentjens R, Rivière I, Park J, Davila M, Wang X, Stefanski J, Taylor C, Yeh R, Bartido S, Borquez-Ojeda O, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–28.CrossRefPubMedPubMedCentral Brentjens R, Rivière I, Park J, Davila M, Wang X, Stefanski J, Taylor C, Yeh R, Bartido S, Borquez-Ojeda O, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–28.CrossRefPubMedPubMedCentral
92.
Zurück zum Zitat Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson DC, Fowler DH, Hardy NM, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–39.CrossRefPubMedPubMedCentral Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson DC, Fowler DH, Hardy NM, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–39.CrossRefPubMedPubMedCentral
93.
Zurück zum Zitat Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73.CrossRefPubMedPubMedCentral Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Ramos C, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, Liu E, Carrum G, Kamble R, Gee A, et al. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains. J Clin Invest. 2016;126:2588–96.CrossRefPubMedPubMedCentral Ramos C, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, Liu E, Carrum G, Kamble R, Gee A, et al. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains. J Clin Invest. 2016;126:2588–96.CrossRefPubMedPubMedCentral
95.
Zurück zum Zitat Turtle C, Hay K, Hanafi L, Li D, Cherian S, Chen X, Wood B, Lozanski A, Byrd J, Heimfeld S, et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of Ibrutinib. J Clin Oncol. 2017;35:3010–20.CrossRefPubMedPubMedCentral Turtle C, Hay K, Hanafi L, Li D, Cherian S, Chen X, Wood B, Lozanski A, Byrd J, Heimfeld S, et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of Ibrutinib. J Clin Oncol. 2017;35:3010–20.CrossRefPubMedPubMedCentral
Metadaten
Titel
Advances on chimeric antigen receptor-modified T-cell therapy for oncotherapy
verfasst von
Yanyu Pang
Xiaoyang Hou
Chunsheng Yang
Yanqun Liu
Guan Jiang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2018
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0840-y

Weitere Artikel der Ausgabe 1/2018

Molecular Cancer 1/2018 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.