Abstract
In recent years, cancer treatment involving adoptive cell therapy with chimeric antigen receptor (CAR)-modified patient’s immune cells has attracted growing interest. Using gene transfer techniques, the patient’s T cells are modified ex vivo with a CAR which redirects the T cells toward the cancer cells through an antibody-derived binding domain. The T cells are activated by the CAR primary signaling and costimulatory domains. Such “second generation” CAR T cells induced complete remission of B cell malignancies in the long-term. In this fast-moving field with a growing number of engineered T cell products, we list about 100 currently ongoing trials here that involve CAR T cells targeting hematopoietic malignancies and solid cancer. Major challenges in the further development of the therapy are briefly discussed.
Keywords: Chimeric antigen receptor, CAR, Clinical trial, Adoptive cell therapy, T cell
Introduction
Over the past few years, adoptive T cell therapy aimed at pre-defined targets has become one of the most promising strategies in cancer immunotherapy, recognized as a “breakthrough therapy” by the US Food and Drug Administration (FDA). T cells were genetically equipped with a chimeric antigen receptor (CAR), which is a composite membrane receptor molecule and provides both targeting specificity and T cell activation (Fig. 1). The CAR targets the T cell through an antibody-derived binding domain in the extracellular moiety, and T cell activation occurs via the intracellular moiety signaling domains when the target is encountered. The CAR design has several advantages over the naturally occurring T cell receptor for antigen (TCR). These include the modular composition of the receptor molecule, redirection toward a broad variety of targets beyond classical MHC-bound peptides, and the combination of signaling domains for sustained T cell activation. Once the CAR engages its cognate target, CAR T cells initiate their immune response with the release of pro-inflammatory cytokines and the cytolytic elimination of the target cell [1]. Activated T cells undergo multiple rounds of amplification and cytolytic attacks, turning them into “serial killers”. To sustain the immune response in the long-term, the primary TCR signal through CD3ξ was combined with a costimulatory signal, mostly CD28 or 4-1BB (CD137), within the same CAR [2, 3] in order to orchestrate a distinct pattern of effector functions. Such “second generation” CARs have proven an important advancement in the development of clinically efficacious T cell therapy. The combination of primary and costimulatory signals sustains distinct T cell functions; the ideal combination of signals seems to be different for the individual T cell subsets and is currently being explored in trials. The 4th generation of CARs contains a CAR inducible expression cassette for a transgene, which may be IL-12 or any other “payload”, thereby making CAR T cells to a targeted factory which delivers a defined transgenic protein to the targeted tissue [4].
Fig. 1.
Families of CARs. a The prototype CAR consists in the extracellular domain of a single chain fragment of variable region (scFv) antibody and a spacer, typically the IgG1 CH2CH3, and in the intracellular domain of the CD3ξ signaling chain providing the primary signal (1st generation CAR). The CAR may harbor one costimulatory or two costimulatory domains in addition to the CD3ξ chain (2nd generation CAR, 3rd generation CAR, respectively). Other signaling chains and various combinations are also described. TRUCK T cells (4th generation) are CAR T cells with CAR inducible release of a “payload” which is a transgenic product, e.g., a cytokine such as IL-12 [4]. b The inhibitory CAR (iCAR) harbors a suppressing instead of an activating signal domain. c Split CAR systems consist of two CARs in the same T cell; the CARs recognize different antigens by their individual scFvs, one CAR providing the CD3ξ signal, the other CAR the costimulatory signal required for full T cell activation (costimulatory CAR) or an suppressor signal to suppress T cell activity (inhibitory CAR). d The switch receptor harbors the PD-1 extracellular domain, or any other receptor for an inhibitory ligand, which is linked to the CD28 costimulatory intracellular domain, thereby switching engagement of an inhibitory ligand into an activating signal
In recent years, procedures have been developed for ex vivo engineering of patient’s T cells with a CAR in good manufacturing practice (GMP) quality and for amplification of the engineered cells to produce clinically relevant numbers for use in adoptive cell therapy. Retro- and lentiviral gene transfer techniques are commonly applied, and DNA and RNA transfer are also used in some trials. Supplementation with IL-7, IL-15 and IL-21 favors the ex vivo expansion of T cells with a less mature phenotype and with improved persistence, along with more potent cytokine release and cytolytic activities. All these activities are thought to be crucial for an efficacious anti-tumor response. In this context, the “pre-conditioning” of the patient’s immune system through non-myeloablative chemotherapy prior to adoptive cell therapy proved to be advantageous in sustaining engraftment and persistence of the transferred CAR T cells. These basic procedures were efficacious in pilot trials, and a number of recent trials are exploring modifications of the basic principle for the treatment of other cancer entities.
CAR T cell trials
As of the beginning of 2016, we are aware of about 100 CAR T cell trials registered at clinicaltrials.gov (Table 1). Most trials have been held in the USA, a growing number is being initiated in Asia and only a few are being carried out in Europe (Table 2). A majority of early-phase trials have been and are currently being performed to treat B cell malignancies, with only a minority of trials targeting solid cancer. Objective regression was achieved in patients with acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL) and various other types of B cell lymphoma upon application of CAR T cells which are redirected against CD19. CD19 is a well-established target in the immunotherapy of B cell malignancies due to its restricted expression by mature B cell lineage cells, but not by other hematopoietic cells or non-hematopoietic tissues. Compared with conventional therapies, including advanced radio- or chemotherapy, CAR T cell trials targeting CD19 exhibited a favorable and lasting clinical outcome. For instance, pediatric and adult patients with ALL experienced a complete remission rate of about 90 % and sustained remission for up to 2 years [5]; 63 % of patients with CLL experienced overall responses and 19 % complete remission [6].
Table 1.
Clinical trials in adoptive cell therapy using second- and third-generation CAR T cells
| Target antigen | Disease | CAR | Gene transfer | T cell origin | Infused dose | Preconditioning | Number of patients | Response | PI | Center | Identifier | References |
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| BCMA | Myeloma | 4-1BB-CD3ξ | NA | Autologous | NA, split dose | Cohen | Abramson Cancer Center | NCT02546167 | ||||
| BCMA | Myeloma | CD28-CD3ξ | Autologous | 0.3–15 × 106 CAR T cells/kg, escalating doses | CTX, FLU | Kochenderfer | NCI | NCT02215967 | ||||
| CD19 | FL | CD28-CD3ξ | RV | Autologous | 108 CAR T cells day 1, 3 × 108 CAR T cells day 2 | CTX, FLU, IL-2 | 1 | 1 × PR | Rosenberg | NCI | NCT00924326 | [35] |
| CD19 | DLBCL, FL | CD3ξ; CD28-CD3ξ | RV | Autologous | 0.2, 1, 2 × 108 CAR T cells/m2 | None | 6 | 2 × SD, 4 × NR | Savoldo | BCM | [7] | |
| CD19 | CLL | CD28-CD3ξ | RV | Autologous | 1.2–3 × 107 CAR T cells/kg, 0.4–1 × 107 CAR T cells/kg, split dose over 2 days |
3 CLL: none 5 CLL: CTX |
8 | 4 × NR, 1 × PR, 2 × SD | Park | MSKCC | NCT00466531 | [14] |
| CD19 | CLL | CD28-CD3ξ | RV | Autologous | 0.3–3 × 107 CAR T cells/kg | CTX, FLU | 4 (8 in total) | 1 × CR, 1 × SD, 2 × PR | Rosenberg | NCI | NCT00924326 | [15] |
| CD19 | CLL | CD28-CD3ξ | RV | Autologous | 1–4 × 106 CAR T cells/kg | CTX, FLU | 4 (15 in total) | 3 × CR, 1 × PR | Rosenberg | NCI | NCT00924326 | [36] |
| CD19 | CLL | CD28-CD3ξ | RV | Allogeneic, donor derived | 1.5, 4.5, 12 × 107 T cells/m2 | None | 4 | 1 × PR, 1 × SD | Ramos | BCM | NCT00840853 | [8] |
| CD19 | Leukemia | CD28-CD3ξ | RV | Allogeneic, donor derived | 0.4–7.8 × 106 CAR T cells/kg | None | 10 | 1 × SD, 2 × NR, 1 × CR | Kochenderfer | NCI | NCT01087294 | [37] |
| CD19 | CLL | Autologous | NA, split dose | CTX, FLU | Hosing | MDACC | NCT01653717 | |||||
| CD19 | CLL, SLL | 4-1BB-CD3ξ | NA | Autologous | 1–5 × 107 CAR T cells, 1–5 × 108 CAR T cells | Frey | Abramson Cancer Center | NCT01747486 | ||||
| CD19 | ALL | 4-1BB-CD3ξ | LV | Autologous | 0.14–1.2 × 107 CAR T cells/kg | 1: None, 1: ETO-CTX | 2 | 2 × CR | Grupp | UPenn | NCT01626495 | [25] |
| CD19 | ALL | 4-1BB-CD3ξ | LV | Autologous | 0.76–20.6 × 106 CAR T cells/kg, split dose | 3: None, 27: physician’s choice salvage chemotherapy | 30 | 27 × CR, 3 × NR | Grupp | UPenn | NCT01626495 | [5] |
| CD19 | ALL | CD28-CD3ξ | RV | Autologous | 0.3, 1, 3 × 107 CAR T cells/kg, split dose over 2 days | CTX | 2 enrolled, 1 treated | 1 × CR | Park | MSKCC | NCT01044069 | [14] |
| CD19 | ALL | CD28-CD3ξ | RV | Autologous | 1.5–3 × 106 CAR T cells/kg, split dose day 1 and 2 | CTX | 5 | 5 × CR | Park | MSKCC | NCT01044069 | [38] |
| CD19 | ALL | CD28-CD3ξ | RV | Autologous | 3 × 106 CAR T cells/kg, 1/3 dose day 1, 2/3 dose day 2 | Physician’s choice salvage chemotherapy and CTX | 16 | 14 × CR, 2 × NR | Park | MSKCC | NCT01044069 | [27] |
| CD19 | ALL | CD28-CD3ξ | RV | Autologous | 1, 3 × 106 CAR T cells/kg | CTX, FLU | 21 enrolled | 14 × CR, 3 × SD, 3 × PD | Lee | NCI | NCT01593696 | [17] |
| CD19 | ALL | CD28-CD3ξ | RV | Allogeneic, donor derived | 1.5, 4.5, 12 × 107 T cells/m2 | None | 4 | 1 × CR, 1 × NR | Ramos | BCM | NCT00840853 | [8] |
| CD19 | ALL | 4-1BB-CD3ξ | LV | Allogeneic, donor derived | NA, split dose | NA | Porter | Abramson Cancer Center | NCT01551043 | |||
| CD19 | ALL | CD28-CD137-CD3ξ | LV | Autologous | NA, split dose | NA | Deng, Hu | Affiliated Hospital to Academy of Military Medical Sciences | NCT02186860 | |||
| CD19 | ALL pediatric | NA | LV | Autologous | NA, split dose | CTX | Gardner | Seattle Children’s Hospital | NCT016832794 | |||
| CD19 | ALL | CD28-CD3ξ | LV | Autologous | NA | Yes | Khaled | COH | NCT02146924 | |||
| CD19 | ALL | CD28-CD3ξ | NA | Autologous | 1, 3 × 106 CAR T cells/kg, split dose | CTX | Curran | MSKCC | NCT01860937 | |||
| CD19 | ALL | 4-1BB-CD3ξ | LV | Autologous | 1–5 × 108 CAR T cells | NA | Frey | Abramson Cancer Center | NCT02030847 | |||
| CD19 | Leukemia | CD28-CD3ξ | RV | Autologous | NA | CTX | Park | MSKCC | NCT01416974 | |||
| CD19 | Leukemia/lymphoma | 4-1BB-CD3ξ | LV | Autologous | 0.146 −16 × 106 CAR T cells/kg, split dose over 3 days | BEN ± RTX; PEN/CTX | 30 | 27 (30) × CR | Frey | Abramson Cancer Center | NCT01029366 | [5, 11, 12] |
| CD19 | Leukemia/lymphoma | CD28-CD3ξ versus CD3ξ | RV | Autologous | 0.2, 1, 2 × 108 CAR T cells/m2 | None | 14 | 14 × no sustained responses | Ramos | BCM | NCT00586391 | [31] |
| CD19 | Leukemia/lymphoma | CD3ξ (EBV-CTLs), CD28-CD3ξ (T cells) | RV | Autologous; syngeneic | 0.2, 1, 2 × 108 CAR T cells/m2 | None | Ramos | BCM | NCT00709033 | [31] | ||
| CD19 | Leukemia | 4-1BB-CD3ξ | LV | Allogeneic, donor derived | NA | Gardner | Seattle Children’s Hospital | NCT02028455 | ||||
| CD19 | Leukemia/lymphoma | 4-1BB-CD3ξ | RV | Autologous; allogeneic | NA, split dose | Han | Chinese PLA General Hospital | NCT01864889 | ||||
| CD19 | NHL | 4-1BB-CD3ξ | NA | Autologous | 1–5 × 108 CAR T cells | Schuster | Abramson Cancer Center | NCT02030834 | ||||
| CD19 | NHL | CD28-CD3ξ | RV | Autologous | 3 × 105 CAR T cells/kg, 1/3 dose day 1, 2/3 dose day 2 | CTX; BEN | Ozawa | Jichi Medical University | NCT02134262 | |||
| CD19 | NHL | CD27-CD3ξ | LV | Autologous | Zhu | Peking University, University of Florida | NCT02247609 | |||||
| CD19 | NHL | CD28-CD3ξ | LV | Autologous | TCM versus TN/MEM | Popplewell | COH | NCT02051257 | ||||
| CD19 | B-cell NHL | CD28-CD3ξ | LV | Autologous | TCM | Popplewell | COH | NCT01815749 | ||||
| CD19 | CLL, NHL, ALL | 4-1BB-CD3ξ | LV | Autologous | NA | Maloney | FHCRC | NCT01865617 | ||||
| CD19 | B-cell NHL, ALL, or CLL | CD28-CD3ξ | RV | Autologous; allogeneic | 0.1, 0.5, 1 × 106 CAR T cells/kg, 0.5, 1, 5 × 106 CAR T cells/kg | Ramos | BCM | NCT02050347 | ||||
| CD19 | MCL | 4-1BB-CD3ξ | RV | Autologous; allogeneic | NA, split dose | Wang | Chinese PLA General Hospital | NCT02081937 | ||||
| CD19 | Stratum 1: NHL stratum 2: CLL, PLL, SLL | CD28-CD3ξ | LV | Autologous | TCM | CTX; BEN; FLU, CTX; ETO, CTX; CTX, ETO | Siddiqi | COH | NCT02153580 | |||
| CD19 | B-cell NHL, ALL, CLL | CD28-CD3ξ, CD28-4-1BB-CD3ξ | RV | Autologous; allogeneic | 1, 5, 20 × 106 CAR T cells/m2 | CTX | Ramos | BCM | NCT01853631 | |||
| CD19 | HL | 4-1BB-CD3ξ | RNA EP | Autologous | NA | Svoboda | Abramson Cancer Center | NCT02277522 | ||||
| CD19 | ALL, CLL, NHL | NA | NA | NA | NA | Qian | Southwest Hospital | NCT02349698 | ||||
| CD19 | leukemia/lymphoma | CD28-CD3ξ | NA | Allogeneic, donor derived | EBV-CTLs, escalating doses | Curran | MSKCC | NCT01430390 | ||||
| CD19 | DLBCL, PMBCL, TFL | NA | NA | Autologous | 2 × 106 CAR T cells/kg | CTX, FLU | Go | Kite Pharma | NCT02348216 | |||
| CD19 | Leukemia/lymphoma | CD28-4-1BB-CD3ξ | RV | Autologous | NA | Yes | Enblad, Hagberg | Uppsala University | NCT02132624 | |||
| CD19 | NHL | CD28-CD3ξ | NA | Autologous | 0.5, 1, 2 × 107 CAR T cells/kg | Carmustine, ETO, Cytarabine, Melphalan | Sauter | MSKCC | NCT01840566 | |||
| CD19 | B-lymphoid malignancies post-HSCT | CD28-CD3ξ | DNA EP | Autologous | NA, split dose | Carmustine, ETO, Cytarabine, Melphalan | Kebriaei | MDACC | NCT00968760 | |||
| CD19 | B-lineage lymphoid malignancies post-UCBT | CD28-CD3ξ | DNA EP | Allogeneic, donor derived | NA | NA | Shpall | MDACC | NCT01362452 | |||
| CD19 | Leukemia/lymphoma | NA | LV | Autologous | CD8+ TCM | NA | Popplewell | COH | NCT01318317 | |||
| CD19 | Leukemia/lymphoma | NA | NA | Allogeneic | CD8+ CMV-CTLs, CD8+ EBV-CTLs | NA | Turtle | FHCRC | NCT01475058 | |||
| CD19 | myeloma | 4-1BB-CD3ξ | LV | Autologous | 1–5 × 107 CAR T cells | Melphalan | 10 | 1 × CR, 3 × PR, 4 × PD | Stadtmauer | Abramson Cancer Center | NCT02135406 | [39] |
| CD19 | Leukemia/lymphoma | NA | NA | NA | NA | NA | Liang | Shanghai Tongji Hospital, Tongji University School of Medicine | NCT02537977 | |||
| CD19 | Leukemia/lymphoma | NA | NA | Autologous | NA | FLU, CTX | Wang | Shenzhen Second People’s Hospital | NCT02456350 | |||
| CD19 | Leukemia/lymphoma | NA | NA | Autologous | 0.5, 1, 3 × 106 CAR T cells/kg | FLU, CTX | Qian | Second Military Medical University | NCT02644655 | |||
| CD19 | Leukemia/lymphoma | NA | NA | Autologous | NA, escalating doses | Yes | Gangyi | Beijing Doing Biomedical Co.; First Hospital of Jilin University | NCT02546739 | |||
| CD19 | Lymphoma | CD28-CD3ξ | RV | Autologous | NA, split dose | Yes | Zhu | Xinqiao Hospital of Chongqing | NCT02652910 | |||
| CD20 | MCL, B-NHL |
CD28-4-1BB- CD3ξ |
DNA EP | Autologous | Infusion with 108, 109, 3.3 × 109 CAR T cells/m2 |
CTX, Aldesleukin |
4 enrolled, 3 treated | 2 × SD, 1 × PR | Till | FHCRC | NCT00621452 | [40] |
| CD20 | Lymphoma | 4-1BB-FcεRIγ | LV | Autologous | NA | Han | Chinese PLA General Hospital | NCT01735604 | ||||
| CD22 |
ALL, FL, NHL, LCL |
4-1BB-CD3ξ | LV | Autologous | 0.3, 1, 3, 10 × 106 T cells/kg | CTX, FLU | Fry | NCI | NCT02315612 | |||
| CD30 | HL, NHL | CD28-CD3ξ | NA | Autologous | 0.2, 1, 2 × 108 T cells/m2 | NA | Ramos | UNC Lineberger CCC | NCT01316146 | |||
| CD30 |
Cutaneous T Cell lymphoma, transformed Mycosis fungoides |
CD28Δ-CD3ξ | RV | Autologous | 1, 3, 10 × 107 CAR T cells, local application | NA | Mauch | University of Cologne | NCT01645293 | |||
| CD30 | HL, NHL | NA | NA | Autologous | escalating doses | CTX, FLU | Han | Chinese PLA General Hospital | NCT02259556 | |||
| CD30 | HL, NHL | CD28-CD3ξ | NA | Autologous | 2, 5, 10 × 108 CAR EBV-CTLs/m2 | NA | Heslop | BCM | NCT01192464 | |||
| CD30 | Lymphoma | NA | LV | NA | NA | Zhu | Peking University | NCT02274584 | ||||
| CD33 | AML | 4-1BB-CD3ξ | LV | Autologous | 1.12 × 109 T cells, escalating doses | 1 | Marked decrease of blasts in bone marrow (2. wk), gradual increase until florid disease progression (9. wk), death (13. wk) | Han | Chinese PLA General Hospital | NCT01864902 | [41] | |
| CD123 | AML | CD28-CD3ξ | LV | Autologous; allogeneic | NA | CTX, FLU, ETO | Budde | COH | NCT02159495 | |||
| CD133 | Leukemia/lymphoma |
CD3ξ; 4-1BB-CD3ξ |
NA | Autologous | NA, escalating doses | Weidong | Chinese PLA General Hospital | NCT02541370 | ||||
| CD138 | Myeloma | 4-1BB-CD3ξ | RV | Autologous; allogeneic | NA | Han | Chinese PLA General Hospital | NCT01886976 | ||||
| LeY | AML | CD28-CD3ξ | RV | Autologous | 5–13 × 108 T cells | FLU | 4 | 3 × clinical responses, 1 × NR | Peter MacCallum Cancer Center | [42] | ||
| LeY | Myeloma, AML, myelodysplastic syndrome | CD28-CD3ξ | RV | Autologous | NA | FLU | Prince | Peter MacCallum Cancer Center | NCT01716364 | |||
| NKG2D-L | Myeloma, AML, myelodysplastic syndrome | NA | NA | NA | 1, 3, 10, 30 × 106 CAR T cells | None | Nikiforow | Celdara Medical, LLC | NCT02203825 | |||
| Igk | Leukemia/lymphoma, myeloma | CD28-CD3ξ | RV | Autologous | 0.2, 1, 2 × 108 CAR T cells/m2 | CTX | Ramos | BCM | NCT00881920 | |||
| ROR1 | CLL, SLL | NA | NA | Autologous | Starting at 105 CAR T cells/kg, escalating doses | FLU, CTX, RTX; BEN, RTX; FLU, BEN, RTX | Wierda | MDACC | NCT02194374 | |||
| CEA | Carcinoma | CD28-CD3ξ | RV | Autologous | NA | Junghans | Roger Williams MC | NCT01723306 | ||||
| CEA | Colorectal cancer | CD28-CD3ξ | RV | Autologous | 0.1, 1, 10 × 1010 T cells | Junghans | Roger Williams MC | NCT00673322 | ||||
| CEA | Breast cancer | CD28-CD3ξ | RV | Autologous | 0.1, 1, 10 × 1010 T cells | Junghans | Roger Williams MC | NCT00673829 | ||||
| CEA | Liver metastases | CD28-CD3ξ | RV | Autologous | 3 infusions in 6 weeks into the hepatic artery, intra-patient dose escalating: 0.1, 1, 10, 30 × 109 CAR T cells | Katz | Roger Williams MC | NCT01373047 | [30] | |||
| CEA | Carcinoma | NA | NA | Autologous | NA | Quian | Southwest Hospital | NCT02349724 | ||||
| CEA | Carcinoma | NA | NA | NA | NA | Quian | Southwest Hospital | NCT02349724 | ||||
| cMet | Breast cancer | NA | RNA EP | Autologous | NA | Tchou | Abramson Cancer Center | NCT01837602 | ||||
| ErbB2 | Carcinoma | NA | Autologous | NA, split dose | Wang | Chinese PLA General Hospital | NCT01935843 | |||||
| ErbB2 | Carcinoma | CD28-CD3ξ | RV | Autologous |
0.01, 0.03, 1, 3, 10, 30, 100 × 106 CAR T cells/m2 |
Gottschalk | BCM | NCT00889954 | ||||
| ErbB2 | Glioblastoma | CD28-CD3ξ | RV | Autologous | 1, 3, 10, 30, 100 × 106 CMV-CTLs/m2 | Ahmed | BCM | NCT01109095 | ||||
| ErbB2 | Sarcoma | CD28-CD3ξ | RV | Autologous |
0.01, 0.03, 1, 3, 10, 30, 100 × 106 CAR T cells/m2 |
CTX, FLU | Ahmed | BCM | NCT00902044 | |||
| ErbB2 | HNSCC | CD28-CD3ξ | RV | Autologous |
107–109 CD4+ T cells, escalating doses |
None | Maher | KCL | NCT01818323 | [43] | ||
| ErbB2 | Carcinoma | NA | NA | Autologous | NA | CTX, FLU, Mesna | Rosenberg | NCI | NCT00924287 | |||
| ErbB2 | Breast cancer | CD28-CD3ξ | RV | Autologous | NA | Yes | Niu | Fuda Cancer Hospital | NCT02547961 | |||
| EGFRvIII | Glioblastoma | NA | LV | Autologous | NA | O’Rourke, Chang | Abramson Cancer Center | NCT02209376 | ||||
| EGFRvIII | Glioma |
CD28-4-1BB- CD3ξ |
RV | Autologous | NA | CTX, FLU, Aldesleukin | Rosenberg | NCI | NCT01454596 | [44] | ||
| EGFR | Glioma | NA | NA | Autologous | NA | Li | RenJi Hospital | NCT02331693 | ||||
| EGFR | Carcinoma | 4-1BB-CD3ξ | LV | Autologous | NA, split dose | Wang | Chinese PLA General Hospital | NCT01869166 | ||||
| EphA2 | Glioma | NA | NA | NA | NA | None | Niu | Fuda Cancer Hospital | NCT02575261 | |||
| FAP | Mesothelioma | CD28-CD3ξ | RV | Autologous | 1 × 106 CD8+ CAR T cells | Stupp | University Hospital Zurich | NCT01722149 | ||||
| FR-α | Ovarian cancer | NA | NA | Rosenberg | NCI | NCT00019136 | ||||||
| GD2 | Sarcoma, melanoma | OX40-CD3ξ | NA | Autologous | 0.1, 1, 3, 10 × 106 CAR T cells/kg | CTX | Kaplan | NCI | NCT02107963 | |||
| GD2 | Neuroblastoma | OX40-CD3ξ | RV | Autologous | 1.5, 2 × 108 CAR T cells | Pembrolizumab, CTX, FLU | Heczey | BCM | NCT01822652 | |||
| GD2 | Neuroblastoma | NA | RV | Allogeneic, donor derived | NA | Myers | Children’s Mercy Hospital Kansas City | NCT01460901 | ||||
| GD2 | Neuroblastoma | CD28-OX40-CD3ξ | RV | Autologous | 0.3, 1, 3, 10 × 107 CAR NK T cells | CTX, FLU | Heczey | BCM | NCT02439788 | |||
| GD2 | Sarcoma |
CD28-OX40- CD3ξ |
NA | Autologous | 0.1, 1, 10 × 107 CAR-VZV-CTLs/m2 | Wang | BCM | NCT01953900 | ||||
| IL-13Ra2 | Glioma | 4-1BB-CD3ξ | LV | Autologous | NA | Badie | COH | NCT02208362 | ||||
| L1-CAM | Neuroblastoma, ganglioneuroblastoma | 4-1BB-CD3ξ; CD28-4-1BB-CD3ξ | LV | Autologous | 0.5, 1, 5, 10 × 106 T cells/kg | Yes | Park | Seattle Children’s Hospital | NCT02311621 | |||
| Mesothelin | Carcinoma | 4-1BB-CD3ξ | LV | Autologous | 1, 3, 10, 30 × 107 CAR T cells/m2 | Haas | Abramson Cancer Center | NCT02159716 | ||||
| Mesothelin | Metastatic cancer | NA | RV | Autologous | NA | CTX, FLU, Aldesleukin | Rosenberg | NCI | NCT01583686 | |||
| Mesothelin | Metastatic pancreatic ductal adenocarcinoma (PDA) | 4-1BB-CD3ξ | RNA EP | Autologous | 1–3 × 108 CAR T cells/m2 (3 times/wk, 3 wks) | Beatty | Abramson Cancer Center | NCT01897415 | ||||
| Mesothelin | Malignant pleural mesothelioma | 4-1BB-CD3ξ | RNA EP | Autologous |
108 CAR T cells, 3 times and 109 CAR T cells, 3 times |
4 | 1 × transient PR, 1 × PD | Haas | Abramson Cancer Center | NCT01355965 | [23, 45] | |
| Mesothelin | Carcinoma | 4-1BB-CD3ξ | NA | Autologous | NA, escalating doses, split dose | Weidong | Chinese PLA General Hospital | NCT02580747 | ||||
| Mesothelin | Pancreas carcinoma | 4-1BB-CD3ξ | LV | Autologous | 1–3 × 107 CAR T cells/m2, 1-3 × 108 CAR T cells/m2, split dose, two separate infusions (Meso CAR T cells and CD19 CAR T cells) | CTX | Ko | UPenn | NCT02465983 | |||
| MUC1 | Carcinoma | CD28-4-1BB-CD3ξ (SM3scFv, IL-12; pSM3scFv) | LV | Autologous | 5 × 105 CAR T cells per tumor lesion, intratumoral injection | None | 1 | pSM3-CAR treated tumor lesion showed necrosis | Yang | PersonGen Biomedicine | NCT02587689 | [46] |
| MUC1 | Carcinoma, glioma | NA | NA | NA | NA | Yang | PersonGen Biomedicine, Anhui General Hospital of Armed Police Forces | NCT02617134 | ||||
| PSMA | Prostate cancer | CD28-CD3ξ | RV | Autologous | 1, 3, 10 × 107 CAR T cells/kg | CTX | Slovin | MSKCC | NCT01140373 | |||
| PSMA | Prostate cancer | NA | RV | Autologous | 1, 10 × 1010 T cells | Junghans | Roger Williams MC | NCT00664196 | ||||
| PSMA | Prostate cancer | NA | RV | Autologous | Junghans | Roger Williams MC | NCT01929239 | |||||
| VEGFR-II | Metastatic cancer, melanoma, renal cancer | NA | RV | Autologous | 1 × 106–3 × 1010 T cells, escalating doses | CTX, FLU, Aldesleukin | Rosenberg | NCI | NCT01218867 |
Trials using second or third generation CAR T cells in the treatment of malignant diseases and registered at http://clinicaltrials.gov as of May 2016 are listed
ALL acute lymphoblastic leukemia, BCM Baylor College of Medicine, BEN bendamustine, CLL chronic lymphocytic leukemia, COH City of Hope Medical Center, CR complete response, CTX cyclophosphamide, DLBCL diffuse large B cell lymphoma, EP electroporation, ETO etoposide, FHCRC Fred Hutchinson Cancer Research Center, FL follicular lymphoma, FLU fludarabine, HL Hodgkin’s lymphoma, LV lentiviral, MC Medical Center, MCL mantle cell lymphoma, MDACC MD Anderson Cancer Center, MSKCC Memorial Sloan Kettering Cancer Center, NA not available, NCI National Cancer Institute, NHL non-Hodgkin lymphoma, NR non-responder, PEN pentostatin, PLL prolymphocytic leukemia, PMBCL primary mediastinal B cell lymphoma, PR partial response, RTX rituximab, RV retroviral, SD stable disease, SLL small lymphocytic lymphoma, TCM central memory T cell, TN/MEM naïve memory T cell, UCBT umbilical cord blood transplantation, UPenn University of Pennsylvania, wk week
Table 2.
World distribution of clinical trials with second- and third-generation CAR T cells
| USA and Canada | 74 |
| PR China | 27 |
| Europe | 4 |
| Japan | 1 |
| Australia | 1 |
The clinical benefit of CAR T cell therapy in these and other trials is well documented. However, the success rate varies broadly between the trial centers and the pathological type of the disease. For instance, 2 out of 6 lymphoma patients achieved stable disease for some months before progression [7], while half the patients (4 out of 8) experienced complete and partial remission in another trial [8]. However, a direct comparison of the outcome is difficult to make due to a number of technical and design variations, including the CAR composition, production and amplification of CAR T cells, patient pre-conditioning and cytokine support, CAR T cell dose and other variations [9]. Lymphodepletion is also a relevant variable in both autologous and allogeneic cell therapy (Table 3). A recent meta-analysis of CD19 CAR T cell trials confirmed lymphodepletion and CAR T cell dose as key factors for a favorable prognosis, while IL-2 co-administration is not recommended [10]. The situation is even more complex when targeting solid cancer lesions. This is due to the fact that, in contrast to CD19, most targeted antigens are likewise expressed by healthy tissues, although often at lower levels. Designed to test for safety, the phase I trials explored the possibility of ruling out the “on-target off-tumor” auto-reactivity of CAR T cells toward healthy tissue which in case of targeting CD19 produced a lasting depletion of healthy B cells. First, phase I trials targeting B cell leukemia/lymphoma have been successfully completed and are currently entering phase II development by global pharmaceutical companies.
Table 3.
Patient pre-conditioning by non-myeloablative lymphodepletion impacts clinical outcome of CAR T cell therapy
| CAR T cells | Pre-conditioning | No pre-conditioning | ||
|---|---|---|---|---|
| Allogeneic | n = 8 |
CR: 2 (25 %) PR: 1 SD: 2 NR: 3 |
||
| Autologous | n = 75 |
CR: 53 (70.7 %) PR: 7 SD: 7 NR: 8 |
n = 24 |
CR: 1 (4.2 %) PR: 14 SD: 2 NR: 7 |
Hematologic malignancies
The first clinical investigations at the University of Pennsylvania with second generation CAR T cells achieved two complete responses in the treatment of three patients with refractory advanced CLL using anti-CD19 CAR T cells [11, 12]; a recent update by the same group confirmed an overall response rate of 57 % [13]. T cells engineered with a “second generation” CAR with combined 4-1BB-CD3ξ signaling underwent extensive amplification upon administration to the patients, eliminated high tumor burdens and persisted for at least 3 years, with retention of anti-tumor activity. With respect to the eliminated tumor mass, it was calculated that one CAR T cell is capable of killing as many as 1000 leukemic cells. At the same time, similar clinical results were obtained by the Memorial Sloan Kettering Cancer Institute [14] and National Cancer Institute [15] using anti-CD19 CAR T cells with combined CD28-CD3ξ signaling. In all trials, the anti-tumor effect correlated with the persistence of CAR T cells in the peripheral blood of the individual patients. Up to now, over 40 trials are targeting CD19 to treat hematologic malignancies (Table 1), including non-Hodgkin’s lymphoma (NHL), CLL and ALL, with other trials targeting CD20 (NCT00621452), CD22 (NCT02315612), Igκ (NCT00881920) and the B cell maturation protein BCMA (NCT02215967).
Larger trials on the treatment of adult and pediatric ALL revealed even more impressive response rates. Complete clinical responses of 90 % were reported in 30 relapsed or refractory ALL patients with ongoing responses over 4 years [5]. Likewise, other trials also obtained complete response rates of 88 and 70 % [16, 17]. A number of patients had previously undergone allogeneic hematopoietic stem cell transplantation, and the T cells from the transplant donor were engineered with the respective CAR. The kinetics of ALL cell clearance in those patients was more rapid compared to that for CLL patients. In addition to an increased sensitivity toward CAR T cells, some further differences in the disease itself may also contribute to the effect, including different tumor burdens, accessibility to immune cells and a more homogeneous patient population treated thus far. In the long-term, most patients treated with 4-1BB-CD3ξ CAR T cells did not receive further treatment, whereas patients treated with CD28-CD3ξ CAR T cells frequently underwent subsequent allogeneic stem cell transplantation. The difference may be biased by the clinical decision to prefer watchful waiting over consolidation of the results, a decision that is based partly on the prolonged persistence of 4-1BB-CD3ξ CAR T cells of up to 2 years, compared to about 30 days for CD28-CD3ξ CAR T cells.
Solid cancer
The application of the CAR T cell strategy to non-hematopoietic cancer requires the consideration of additional factors. These include disease status and tumor burden, immune repression in the tumor tissue and CAR T cell infiltration, as well as the recruitment and activation of other pro-inflammatory and repressor immune cells. Some early trials are currently investigating the safety and potential side effects of CAR T cell-targeting of solid cancer lesions after systemic application, while other trials apply CAR T cells through local administration. Not all effective variables for therapeutic efficacy have as yet been identified; among these, the choice of target antigen is a major issue. Redistributed expression of an antigen throughout the entire membrane of cancer cells, while polarized expression in glandular or intestinal epithelia, makes such an antigen targetable by CAR T cells. These targets include prostate-specific membrane antigen (PSMA), carcinoembryonic antigen (CEA) and mucin 1 (Muc1), all currently being investigated in clinical trials. Two trials using anti-CEA CAR T cells are currently recruiting (NCT00673829, NCT01723306); one trial was terminated due to treatment-related adverse effects (NCT01212887).
CAR T cell therapy is a tightrope walk
Although adoptive CAR T cell therapy achieved spectacular efficacy in the treatment of leukemia/lymphoma, this treatment is often associated with significant toxicities which need to be taken into account, in particular, when entering advanced phases of clinical evaluation.
-
Most toxicities in CAR T cell therapy are due to the engagement of the cognate antigen on healthy cells. Such “on-target off-tumor” targeting may harbor life-threatening risks, in particular, when the antigen is expressed by essential tissues such as lung, heart or liver. In the case of anti-CD19 CAR T cells for the treatment of B cell leukemia/lymphoma, lasting B cell aplasia and, consequently, hypo-gammaglobulinemia are consistently observed, which is considered a biomarker for anti-CD19 CAR T cell function. The situation can be clinically managed by immunoglobulin substitution, and patients generally did not develop opportunistic infections. To allow reconstitution of the B cell compartment, CAR T cell elimination after complete tumor eradication would be ideal which can be achieved by various means as described below.
The severity of “on-target off-tumor” toxicity became obvious after the death of a patient during the treatment of metastatic colon cancer (NCT00924287), when CAR T cells targeted HER2/neu (ErbB2), which is highly expressed by the carcinoma cells but also by healthy tissue, although at lower levels, causing acute toxicity toward cardiopulmonary epithelia [18]. Other trials are currently exploring anti-HER2 CAR T cells in the treatment of brain tumors and various carcinomas (NCT00889954, NCT00902044, NCT01818323, NCT01935843). Despite the risk of toxicity, anti-HER2 CAR T cells can be safe when using a less potent signaling CAR and applying a more cautious dose-escalation regimen [19].
In cases of toxicity, CAR T cells can be eliminated by the activity of co-expressed inducible suicide genes (herpes simplex virus thymidine kinase, HSV-tk; inducible caspase 9, iCasp9), by the administration of a clinically approved depleting antibody targeting an epitope within the CAR, or a co-expressed non-functional surface protein, all strategies that are being explored in trials. For instance, HSV-tk, which phosphorylates the guanosine analog ganciclovir which initiates the arrest of DNA replication, is used in trials targeting PSMA in prostate cancer (NCT01140373), CD19 in NHL (NCT00182650) and L1-CAM in neuroblastoma (NCT00006480). Inducible caspase 9, which encodes a modified truncated caspase 9 and initiates an apoptotic cascade upon drug-initiated dimerization, is being explored in trials targeting the disialoganglioside GD2 in neuroblastoma (NCT01822652), sarcoma (NCT01953900), other solid cancers (NCT02107963) and in trials targeting CD19 for the treatment of NHL (NCT02247609). The truncated EGFR is co-expressed with the CAR in the same T cell and targeted by the anti-EGFR therapeutic antibody cetuximab in various trials (NCT01815749, NCT02051257).
The level of antigen expression is clearly a critical factor for “off-tumor” auto-immunity. Moreover, the CAR T cell strategy demands a target which is exclusively expressed by cancer cells, ideally required for their survival and harboring mutations that are large enough to produce new epitopes that can be specifically recognized by the CAR [20]. In the absence of such an ideal target, however, target antigens are preferred that are either co-expressed by non-essential tissues or topologically sequestered from T cells. In the latter case, epithelial antigens with polarized membrane distribution in healthy cells, while uniformly distributed on cancer cells, like CEA, provide some tumor selectivity based on altered topology.
Several strategies are currently being developed to furthermore increase tumor selectivity. These strategies include the co-expression of inhibitory CARs (iCARs) with PD-1 or CTLA-4 intracellular domains and targeting a surface antigen on healthy tissues which is not present on cancer cells, by an inhibitory CAR thereby providing a dominant inhibitory signal when engaging healthy cells (Fig. 1) [21]. Alternatively, the CAR T cell activation may depend on the CAR engagement of antigen pairs, where only binding to both antigens by both CARs drives full T cell activation [22]. Clinical exploration of the latter strategy remains challenging since it requires the adjustment of CARs to the individual antigen levels in tumors and healthy tissues.
In this situation, T cells which are transiently modified by a CAR, thereby limiting the persistence and function of CAR T cells, were proposed. RNA-modified CAR T cells require repetitive CAR T cell infusions as applied in a trial that targeted mesothelioma (NCT01355965) [23]. Other trials are using RNA-modified CAR T cells against pancreatic cancer (NCT01897415), breast cancer (NCT01837602) and Hodgkin’s lymphoma (NCT02277522).
-
CAR T cell activation is accompanied by extensive release of toxic levels of pro-inflammatory cytokines which can cause cytokine release syndrome (CRS), characterized by nausea, fever, hypotension, vascular leakage and life-threatening multiple organ failure. This syndrome is closely associated with systemic macrophage activation syndrome, resembling hematophagocytic lymphohistiocytosis. CRS is correlated with clinical efficacy. The pathomechanism seems to be based on the extensive activation of CAR T cells, with high IFN-γ and TNF-α release and the activation of monocytes or macrophages, which release IL-6, which in turn produces a toxic effect on various organs such as kidney, liver and brain. Major risks for CRS are therefore thought to be a high tumor burden and the dose and potency of applied T cells. However, patients with low tumor loads also suffered from CRS. The severity of CRS requires intensive care treatment, and the death of two cancer patients in 2014 has demonstrated that CRS constitutes a severe limitation of CAR T cell therapy [15, 24, 25]. Interference with the IL-6 pathway through application of the neutralizing anti-IL-6 antibody tocilizumab successfully reduced the symptoms without eliminating CAR T cells [26]. In order to clinically monitor CRS and to grade the severity of CRS, a clear treatment algorithm has recently been established based on tumor burden, age, comorbidities and other factors [27, 28].
Acute and reversible neurotoxicity after CD19 CAR T cell application were observed in about 40 % of patients during CRS, with 20 % of patients experiencing late-onset manifestations with aphasia, hallucinations, and delirium. Adult patients and children equally suffered from CRS [29]. While the exact pathomechanism remains unclear and only partly overlaps with CRS, there is some evidence that CAR T cells may infiltrate the brain parenchyma, induce diffuse encephalopathy and release IL-6, which is not neutralized by i.v. application of tocilizumab.
Alternative routes for the delivery of T cells beyond i.v. infusion are also being explored to minimize the risk of “off-tumor” toxicities. In particular, the CAR T cells were administered in a localized fashion by intratumoral (CEA, NCT01373047; HER2, NCT01818323), intracerebral (EGFRvIII, NCT01454596) or intrapleural (mesothelin, NCT01355965) injections. Individual solid cancer metastases were treated by locally introducing the CAR T cells into the tumor lesion. For instance, endoscopic delivery of CAR T cells to CEA+ metastases in the liver (NCT01373047) resulted in shrinkage of metastases without systemic side effects [30]. Likewise, mesothelin-specific CAR T cells were introduced into the pleura of mesothelioma patients [23]. Local application of the CAR T cells is assumed to have some advantages, including the prompt antigen-induced activation to sustain robust T cell amplification, persistence in the tumor lesion and the low dose of CAR T cells required to induce efficacy when compared to systemic infusion. At present, some clinical trials have incorporated such tools for minimizing potential systemic toxicities.
Once accumulated at the tumor site, the CAR T cell anti-tumor response is repressed by various means, for instance, by preventing escape from the vasculature and penetration into the tumor tissue, repression by repressor cells and/or soluble factors, or by nutrient deprivation. Moreover, CAR T cells express immune repressive receptors upon activation, including programmed cell death-1 (PD-1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). Pre-clinical models have demonstrated strategies to counteract the PD-1 or CTLA-4-mediated CAR T cell repression, while the effect of the co-administered anti-CTLA-4 antibody, ipilimumab, is currently being investigated in a pilot trial (NCT00586391) [31].
Future perspectives
While CAR T cell therapy for refractory leukemia/lymphoma is becoming more and more established in clinical practice, CAR T cell therapy for solid cancer is still in its infancy and requires extensive clinical investigation [32]. Moreover, the comparison of trial results is complicated by a number of differences in CAR T cell formulation, trial protocols, pre-conditioning of patients and other factors. The current situation demands a more rigorous standardization; some major obstacles need attention in the near future.
Target selection: Most targeted antigens are tumor-associated but not tumor-selective as tumor-associated antigens are also expressed by healthy tissues, making them potential CAR T cell targets. In extensive pre-clinical studies and sophisticated mouse models, researchers are trying to define the risk of CAR-mediated immunity against healthy tissues. However, the limitations of such models demand a responsible design of early-phase trials. It more and more becomes obvious that CAR T cells can be safe when targeting with less affinity the appropriate target and with less signaling capacities. Translation to clinical application still demands a cautious dose-escalation regimen and a rigorous clinical monitoring. In this respect, trials are aiming at minimizing the “off-tumor” risk while increasing CAR T cell efficacy toward cancer cells. As a consequence, engineered T cells, which recognize target antigen signatures, instead of an individual antigen, indicative for cancer cells by split receptor systems (Fig. 1), are experimentally explored. Two CARs are co-expressed in T cells complementing their activating signals when engaging both antigens, thereby increasing cancer selectivity and likely improving therapeutic safety. In a further development, a CAR with extracellular PD-1 domain, which binds to PD-L1 and PD-L2, provides CD28 co-stimulation as the required second signal in an immune suppressive tumor environment with PD-1 ligand expression [33]. Such split CARs can also be used to provide inhibitory signals to the T cell when targeting an antigen on healthy tissues.
CAR design and T cell formulation: Different CAR designs with respect to the extracellular spacer, transmembrane domain and costimulatory moieties are used; most modifications are merely empiric or were found functional in the particular context, others still need systemic evaluation. So far, the binding affinity, the targeted epitope and the extracellular length of the CAR were identified to be crucial for optimal T cell activation. Some optimization applies for the ex vivo amplification of engineered T cells to clinically relevant numbers, which is performed in the presence of IL-2, now preferentially in the presence of IL-7 and IL-15. In addition, the T cell subset undergoing CAR modification substantially impacts clinical outcome. Standardization of the manufacturing processes will help to obtain cell products which allow comparing the clinical performance in different trials on a more robust basis. Currently available clinical data suggest that CAR T cells with 4-1BB-ξ signaling domain inducing a central memory phenotype persist longer than T cells with CD28-ξ CAR inducing an effector memory phenotype. Moreover, a recent analysis points to differences in the metabolic reprogramming by 4-1BB-ξ versus CD28-ξ CARs to central versus effector T cells, respectively [34]. However, other T cell subsets and T cells in a more advanced stage of maturation need other costimulatory signals or combinations thereof to persist in the long-term. Until now, autologous and allogeneic CAR T cells were applied with different outcome; however, there are too many differences in the trial and CAR design which do not allow a well-founded trial comparison. Future efforts should be focused on a series of comparative trials with the aim to identify the optimal design.
Pre-conditioning: CAR T cell amplification after application is clearly required for an anti-tumor response; non-myeloablative lymphodepletion of patients prior T cell therapy is crucial in this context (Table 3). The additional impact of pre-conditioning on sensitizing the tumor milieu by depleting suppressor cells or inducing the release of antigen needs to be evaluated; however, the pre-treatment protocols and their efficacies substantially differ and need further standardization. In addition, the optimal dose of IL-2 substitution during and shortly after T cell therapy in order to sustain T cell amplification and the use of alternative cytokines such as IL-7 and/or IL-15 need to be clinically explored.
Control of side effects: The cytokine release and the vascular leakage syndromes are severe, life-threatening side effects which seem to be associated with anti-tumor activity of CAR T cells but need intensive care attention. With the establishment of a CRS screening protocol and IL-6 neutralization [27–29], first steps toward a more standardized clinical management of those side effects of the CAR T cell therapy are being made.
Hematopoietic stem cell transplantation: When CAR T cell therapy has induced clinical remission, a number of patients were transplanted with allogeneic hematopoietic stem cells. While the anti-leukemia efficacy of CAR T cells is clearly shown, the capacity to control leukemia in the long-term without stem cell transplantation needs to be established in forthcoming trials.
Acknowledgments
Work in the authors’ laboratory was supported by the Deutsche Forschungsgemeinschaft, Bonn, Germany, the Deutsche Krebshilfe, Bonn, Germany, the Deutsche José Carreras-Leukämie Stiftung, Munich, Germany, the Else Kröner-Fresenius Stiftung, Bad Homburg v.d.H., Germany, and the Fortune Program of the Medical Faculty of the University of Cologne, Cologne, Germany.
Abbreviations
- ALL
Acute lymphocytic leukemia
- CEA
Carcinoembryonic antigen
- CLL
Chronic lymphocytic leukemia
- CRS
Cytokine release syndrome
- NHL
Non-Hodgkin’s lymphoma
- PSMA
Prostate-specific membrane antigen
- tk
Thymidine kinase
Compliance with ethical standards
Conflict of interest
Hinrich Abken serves on the scientific advisory board of Miltenyi Biotec. The other authors declare no conflict of interest.
References
- 1.Eshhar Z. The T-body approach: redirecting T cells with antibody specificity. Handb Exp Pharmacol. 2008;181:329–342. doi: 10.1007/978-3-540-73259-4_14. [DOI] [PubMed] [Google Scholar]
- 2.Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol. 1998;161:2791–2797. [PubMed] [Google Scholar]
- 3.Hombach A, Wieczarkowiecz A, Marquardt T, et al. Tumor-specific T cell activation by recombinant immunoreceptors: CD3 zeta signaling and CD28 costimulation are simultaneously required for efficient IL-2 secretion and can be integrated into one combined CD28/CD3 zeta signaling receptor molecule. J Immunol. 2001;167:6123–6131. doi: 10.4049/jimmunol.167.11.6123. [DOI] [PubMed] [Google Scholar]
- 4.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 Res. 2011;71:5697–5706. doi: 10.1158/0008-5472.CAN-11-0103. [DOI] [PubMed] [Google Scholar]
- 5.Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Singh N, Frey NV, Grupp SA, Maude SL. CAR T Cell Therapy in Acute Lymphoblastic Leukemia and Potential for Chronic Lymphocytic Leukemia. Curr Treat Options Oncol. 2016;17:28. doi: 10.1007/s11864-016-0406-4. [DOI] [PubMed] [Google Scholar]
- 7.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 Investig. 2011;121:1822–1826. doi: 10.1172/JCI46110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Cruz CRY, Micklethwaite KP, Savoldo B, et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood. 2013;122:2965–2973. doi: 10.1182/blood-2013-06-506741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Xu X-J, Zhao H-Z, Tang Y-M. Efficacy and safety of adoptive immunotherapy using anti-CD19 chimeric antigen receptor transduced T-cells: a systematic review of phase I clinical trials. Leuk Lymphoma. 2013;54:255–260. doi: 10.3109/10428194.2012.715350. [DOI] [PubMed] [Google Scholar]
- 10.Zhang T, Cao L, Xie J, et al. Efficiency of CD19 chimeric antigen receptor-modified T cells for treatment of B cell malignancies in phase I clinical trials: a meta-analysis. Oncotarget. 2015;6:33961–33971. doi: 10.18632/oncotarget.5582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Porter DL, Hwang W-T, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7:303ra139. doi: 10.1126/scitranslmed.aac5415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Brentjens RJ, Rivière I, Park JH, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–4828. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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. doi: 10.1182/blood-2011-10-384388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Davila ML, Bouhassira DCG, Park JH, et al. Chimeric antigen receptors for the adoptive T cell therapy of hematologic malignancies. Int J Hematol. 2014;99:361–371. doi: 10.1007/s12185-013-1479-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lee DW, Kochenderfer JN, Stetler-Stevenson M, 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;385:517–528. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Morgan RA, Yang JC, Kitano M, et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–851. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ahmed N, Brawley VS, Hegde M, 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;33:1688–1696. doi: 10.1200/JCO.2014.58.0225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gross G, Eshhar Z. Therapeutic potential of T cell chimeric antigen receptors (CARs) in cancer treatment: counteracting off-tumor toxicities for safe CAR T cell therapy. Annu Rev Pharmacol Toxicol. 2016;56:59–83. doi: 10.1146/annurev-pharmtox-010814-124844. [DOI] [PubMed] [Google Scholar]
- 21.Fedorov VD, Themeli M, Sadelain M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med. 2013;5:215ra172. doi: 10.1126/scitranslmed.3006597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kloss CC, Condomines M, Cartellieri M, et al. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2012;31:71–75. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Beatty GL, Haas AR, Maus MV, et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res. 2014;2:112–120. doi: 10.1158/2326-6066.CIR-13-0170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Brentjens R, Yeh R, Bernal Y, et al. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial. Mol Ther. 2010;18:666–668. doi: 10.1038/mt.2010.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.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. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chen F, Teachey DT, Pequignot E, et al. Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CAR T cell therapy. J Immunol Methods. 2016;434:1–8. doi: 10.1016/j.jim.2016.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra25. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 2014;20:119–122. doi: 10.1097/PPO.0000000000000035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 2016;6:664–679. doi: 10.1158/2159-8290.CD-16-0040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Katz SC, Burga RA, McCormack E, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric antigen receptor-modified t-cell therapy for CEA + Liver Metastases. Clin Cancer Res. 2015;21:3149–3159. doi: 10.1158/1078-0432.CCR-14-1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Xu Y, Zhang M, Ramos CA, 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;123:3750–3759. doi: 10.1182/blood-2014-01-552174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Abken H. Adoptive therapy with CAR redirected T cells: the challenges in targeting solid tumors. Immunotherapy. 2015;7:535–544. doi: 10.2217/imt.15.15. [DOI] [PubMed] [Google Scholar]
- 33.Kobold S, Grassmann S, Chaloupka M, et al. Impact of a new fusion receptor on PD-1-mediated immunosuppression in adoptive T cell therapy. J Natl Cancer Inst. 2015 doi: 10.1093/jnci/djv146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kawalekar OU, O’Connor RS, Fraietta JA, et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 2016;44:380–390. doi: 10.1016/j.immuni.2016.01.021. [DOI] [PubMed] [Google Scholar]
- 35.Kochenderfer JN, Wilson WH, Janik JE, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116:4099–4102. doi: 10.1182/blood-2010-04-281931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33:540–549. doi: 10.1200/JCO.2014.56.2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kochenderfer JN, Dudley ME, Carpenter RO, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–4139. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted t cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra38. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Garfall AL, Maus MV, Hwang W-T, et al. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N Engl J Med. 2015;373:1040–1047. doi: 10.1056/NEJMoa1504542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Till BG, Jensen MC, Wang J, 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;119:3940–3950. doi: 10.1182/blood-2011-10-387969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang Q, Wang Y, Lv H, et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther. 2015;23:184–191. doi: 10.1038/mt.2014.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ritchie DS, Neeson PJ, Khot A, et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol Ther. 2013;21:2122–2129. doi: 10.1038/mt.2013.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.van Schalkwyk MCI, Papa SE, Jeannon J-P, et al. Design of a phase I clinical trial to evaluate intratumoral delivery of ErbB-targeted chimeric antigen receptor T-cells in locally advanced or recurrent head and neck cancer. Hum Gene Ther Clin Dev. 2013;24:134–142. doi: 10.1089/humc.2013.144. [DOI] [PubMed] [Google Scholar]
- 44.Morgan RA, Johnson LA, Davis JL, et al. Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther. 2012;23:1043–1053. doi: 10.1089/hum.2012.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Maus MV, Haas AR, Beatty GL, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1:26–31. doi: 10.1158/2326-6066.CIR-13-0006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.You F, Jiang L, Zhang B, 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;59:386–397. doi: 10.1007/s11427-016-5024-7. [DOI] [PubMed] [Google Scholar]

