Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2799 2023-03-31 12:47:46 |
2 format -1 word(s) 2798 2023-04-03 03:39:24 | |
3 format -1 word(s) 2797 2023-04-21 08:06:32 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ren, A.; Tong, X.; Xu, N.; Zhang, T.; Zhou, F.; Zhu, H. CAR T-Cell Immunotherapy Treating T-ALL. Encyclopedia. Available online: https://encyclopedia.pub/entry/42691 (accessed on 27 July 2024).
Ren A, Tong X, Xu N, Zhang T, Zhou F, Zhu H. CAR T-Cell Immunotherapy Treating T-ALL. Encyclopedia. Available at: https://encyclopedia.pub/entry/42691. Accessed July 27, 2024.
Ren, Anqi, Xiqin Tong, Na Xu, Tongcun Zhang, Fuling Zhou, Haichuan Zhu. "CAR T-Cell Immunotherapy Treating T-ALL" Encyclopedia, https://encyclopedia.pub/entry/42691 (accessed July 27, 2024).
Ren, A., Tong, X., Xu, N., Zhang, T., Zhou, F., & Zhu, H. (2023, March 31). CAR T-Cell Immunotherapy Treating T-ALL. In Encyclopedia. https://encyclopedia.pub/entry/42691
Ren, Anqi, et al. "CAR T-Cell Immunotherapy Treating T-ALL." Encyclopedia. Web. 31 March, 2023.
CAR T-Cell Immunotherapy Treating T-ALL
Edit

T-cell acute lymphoblastic leukemia (T-ALL), a form of T-cell malignancy, is a typically aggressive hematological malignancy with high rates of disease relapse and a poor prognosis. Guidelines do not recommend any specific treatments for these patients, and only allogeneic stem cell transplant, which is associated with potential risks and toxicities, is a curative therapy. Clinical trials showed that immunotherapies, including monoclonal antibodies, checkpoint inhibitors, and CAR T therapies, are successful in treating hematologic malignancies. CAR T cells, which specifically target the B-cell surface antigen CD19, have demonstrated remarkable efficacy in the treatment of B-cell acute leukemia, and some progress has been made in the treatment of other hematologic malignancies. However, the development of CAR T-cell immunotherapy targeting T-cell malignancies appears more challenging due to the potential risks of fratricide, T-cell aplasia, immunosuppression, and product contamination.

immunotherapy T-ALL CAR T fratricide

1. Fratricide and Promising Strategies

Fratricide, which is caused by the same antigen being expressed on both leukemia cells and Chimeric antigen receptor T-cell (CAR T) cells, is a core problem in the development of therapies for T-cell leukemia/lymphoma, especially for T-cell acute lymphoblastic leukemia (T-ALL). Since T-ALL is derived from normal T cells, it retains a very similar antigen expression as normal T cells, including CD7 and CD5 [1][2][3][4]. This shared expression of antigens can make CAR T cells commit fratricide, limiting the proliferation of CAR T cells in vitro and impeding cell infusion for patients. To inhibit antigen-driven fratricide, several different strategies based on the cell type or antigen have been investigated.

1.1. Transduce CAR beyond T Cells

Because most antigens targeted for T-cell malignancies are not shared with NK cells, and NK cells also exhibit rapid and strong cytotoxicity against tumor cells [5][6], NK cells can be adapted as allogenic CAR-modified cells to overcome the difficulty of autologous CAR T production, without the risk of GvHD given their MHC-independent activation, especially for aggressive T-ALL at relapse [7][8]. Several preclinical studies have been conducted using CAR-modified NK cells or NK-92 cell lines for the treatment of T-cell malignancies, which showed specific cytotoxicity and prolonged survival in vitro in mouse models [9][10]. Two groups reported that a specific CAR NK framework containing 2B4 (CD244) to target CD3, CD4, and CD5 had superior antitumor activity compared with the CAR T framework on NK effector cells in T-cell malignancies [11][12]. Furthermore, anti-CD7 CARs have been developed to treat T-cell malignancies with promising preclinical results, whereas effector T cells modified with anti-CD7 CARs caused extensive fratricide and prevented T-cell expansion [4][13]. Several clinical trials using CAR NK cells instead of CAR T cells for the treatment of CD7-positive T-ALL and T-cell lymphoma have been established (NCT04934774, NCT04840875) [5].
However, NK cells are difficult to expand ex vivo and transduce with viral vectors. NK-92-based CAR products could be developed as a targeted allogeneic cell therapeutic agent. This therapy requires irradiation before infusion into the patient to prevent permanent engraftment [14]. Pinz et al. constructed a CD4 CAR on NK-92 cell lines that specifically eliminated diverse CD4+ human T-cell leukemia/lymphoma cell lines and patient samples ex vivo and efficiently targeted and killed KARPAS-299 cells in vivo [15]. In addition to replacing NK cells with NK-92 cell lines, several methods have been developed to enhance NK cell expansion ex vivo. Kweon et al. created a new effective approach for the ex vivo expansion of human NK cells using K562 cells that were genetically engineered (GE) to express the OX40 ligand (K562-OX40L) in conjunction with brief exposure to soluble IL-21, which enhanced NK cell expansion approximately 2000-fold after 4 weeks of culture [16].

1.2. Find Specific Antigens Restricted Expression on T Cells

Identifying tumor-specific antigens that restrict T-cell expression is another strategy to avert fratricide and overcome CAR T therapy for T-ALL. Currently, TRBC1/2, CDR3, CD1a, CD4, CCR7, CCR9, CD33, CD30, and CD99 are now being identified and evaluated as targets for CAR T therapy to treat T-cell malignancies [9][17][18][19][20][21][22][23]. These biomarkers have limited expression on normal T cells and antigen-derived CAR T cells continue to proliferate normally.
Normal T cells express either TRBC1 or TRBC2, which is coded by the T-cell receptor β-chain constant region [24]. According to normal mature T cells containing TRBC1 and TRBC2, malignancies are restricted to only one positive subtype [25]. Maciocia et al. developed anti-TRBC1 CAR T cells that recognized TCBR1-positive malignant and normal T cells but did not kill TRBC2-positive normal T cells [25]. TRBC-targeted immunotherapy was shown to eradicate a fraction of TCBR1-positive T-ALL while preserving sufficient normal T cells to maintain cellular immunity. Additionally, an ongoing clinical trial is examining the safety and efficacy of TRBC1 CAR T-cell therapy for patients with relapsed/refractory TRBC1 positive T-cell hematological malignancies (NCT04828174).
In addition, each patient has a unique T-cell receptor (TCR) on their leukemia cell surface, which could distinguish these cells from normal T cells. CDR3, the complementarity determining region 3 on TCRs, is very variable and has been used as a target for vaccines to induce antibody response [9]. Huang et al. chose unique CDR3 regions from patients with T-cell leukemia and validated the efficacy and safety of clonal T-cell CDR3-selective CAR T therapy in vitro and in vivo [9]. The CDR3-selective CAR T cells exhibited lower on-target off-tumor effects and minimized the impact on CAR T-cell amplification [9].
CD33, which is expressed on myeloid cells, is commonly used as a target of immunotherapy for AML [26][27]. Moreover, CD33 is also expressed abnormally in 10–20% of B- and T-lymphoblastic leukemias/lymphomas but not in the normal T cells. It is especially highly expressed in the subtype of early T precursor acute lymphoblastic leukemias/lymphoma (ETP-ALL) which is associated with high rates of intrinsic treatment resistance [28][29]. Guo et al. reported that CD33 was expressed in T-ALL and that CD33 highly expressed patients had a poor prognosis [30]. These results suggest that CD33 is a potential target for immunotherapy in ETP-ALL patients. However, there were not sufficient data to support the premise that targeting CD33 therapy was efficient in non-ETP T-ALL.
CCR9, a G-protein-coupled receptor for the natural ligand CCL25, is expressed on less than 5% of normal circulating T cells. Maciocia, P.M. et al. constructed anti-CCR9 CAR T cells, using a novel rat-derived anti-CCR9 scFv, and demonstrated that CCR9 CAR cells efficiently eliminated CCR9 positive tumor cells with no fratricide in vitro and in vivo [31].
Additionally, FACS analysis of T-ALL cell lines showed that CD21 existed on 70% of human T-ALL cell lines, and CD21 positivity varied by maturational stage, with cortical T-ALL exhibiting the highest expression (80% of cases), followed by pre-T (72%), mature (67%), ETP (25%), and pro-T (17%), suggesting that CD21 is a potential target of T-ALL [32]. Maciocia, N.C. et al. reported that CD21, which exhibits minimal expression on mature T cells, is a novel target for CAR T-cell therapy in T-ALL [32]. They successfully demonstrated the efficient treatment of anti-CD21 CAR in murine models of T-ALL.
The antigens mentioned above essentially solve the problems related to fratricide in CAR T immunotherapy. However, CAR T therapy targeting these antigens has not been widely assessed in clinical trials and these antigen-positive hematologic tumors may not cover all T-cell malignancies. More specific antigens remain to be identified.

1.3. Knock-Out Pan-T-Cell Targeting Antigens on CAR T Cells by CRISPR-Cas9

Genome editing techniques including TALEN, zinc-finger nucleases (ZFNs), and CRISPR/Cas9 are used to precisely modify DNA within a cell. In 2018, Rasaiyaah et al. used transcription activator-like effector nuclease (TALEN) mRNA to disrupt the TRAC locus, showing robust cytotoxicity of anti-CD3 CAR T cells against CD3-positive cells [33]. Similarly, CRISPR/Cas9 has been adapted to disrupt endogenous genes in CAR T therapy to prevent graft-versus-host disease (GvHD) [34]. Furthermore, another strategy for preventing fratricide is using CRISPR-Cas9 gene editing technology to knock out the Pan-T-cell surface antigen expressed on CAR T cells, such as CD3, CD5, and CD7, which are also present in healthy T cells [6][13][35]. For example, a preclinical study demonstrated that gene editing of CD5 in effector CAR T cells using CRISPR-Cas9 gene editing technology significantly increased the expression of CAR and the antitumor activity of anti-CD5 CAR T cells [6]. CD7, as a CAR target for T-ALL, was shown to exhibit extensive fratricide. Thus, several groups used the CRISPR/Cas9 system to disrupt the CD7 locus and demonstrated that anti-CD7 CAR T cells retained antitumor activity without killing gene-disrupted T cells in preclinical and clinical studies (NCT04264078 and NCT04004637). Recently, Hu et al. reported encouraging results from a phase 1 clinical trial (NCT04538599) in which 11 patients with refractory/relapse T-cell leukemia/lymphoma and one with CD7 positive acute myeloid leukemia received treatment of genetically modified CD7-targeting allogeneic CAR T cells.
CRISPR/Cas9-mediated editing of CAR T cells could be used to overcome CAR T-cell fratricide by targeting the antigens expressed on them. However, CRISPR/cas9 may cause DNA double-stranded breaks (DSBs), resulting in other genes being knocked out, with potentially unforeseen consequences, such as complex genomic rearrangements, megabase-scale deletions, and chromothripsis [36][37]. Recently, Diorio et al. created cytosine base editor (CBE) technology, which has more than 90% efficiency in terms of gene silencing due to point mutation without inducing DNA double-stranded breaks. She applied CBEs to create CD7-directed allogeneic CAR T using four simultaneous base edits. The CBE-edited CAR T cells exhibited normal proliferation and were shown to be highly efficient for T-ALL treatment in vitro and in vivo [38].
Other non-gene editing strategies have been explored to overcome CAR T fratricide. For instance, Mamonkin et al. designed a Tet-OFF expression system in which CAR expression controlled by doxycycline was imported to halt fratricide among CD5-41BB -CAR T cells in vitro and in vivo [37]. Furthermore, Png et al. used protein expression blockers (PEBLs) to maintain the targeted protein in the endoplasmic reticulum (ER)/golgi, which stopped the antigen expressing on the cell surface of CAR T cells by combining an scFv with a retention peptide. This exhibited progressively increased cytotoxicity against T-ALL blasts in vitro and significant survival advantages in vivo compared with the control group [4]. Similarly, anchoring CD7 in the ER and/or golgi to overcome CAR T fratricide was assessed in a clinical study in which all three patients were controlled (NCT04004637) [39]. In addition, Pan et al. applied donor-derived CD7 CAR T cells in a phase 1 trial for 20 r/r T-ALL patients (NCT04689659, ChiCTR2000034762) [40]. They designed CD7 CAR T without surface expression of CD7 using IntraBlock technology and reserved intracellular expression by constructing the CD7 binding domain in the vector, which could be fused with an ER retention signal sequence (KDEL) [41]. The trial achieved desirable results in which 18 patients exhibited complete remission with reversible adverse events, despite a patient dying through pulmonary hemorrhage related to fungal pneumonia after 5.5 months of transfusion.

2. T-Cell Aplasia and Proposed Solutions

T-cell aplasia, which is caused by on-target off-tumor toxicity of CAR T cells against normal T cells with the expression of CAR-specific target antigens, is another major obstacle to the development of CAR T cells for T-cell malignancies [42]. CAR T cells directed against such targets, which are generally expressed in normal T cells, lead to profound life-threatening infections. However, several strategies have been found to avoid the incidence of T-cell aplasia.
Selecting the specific antigen of malignant T cells (no expression or limited expression on normal T cells) is an alternative method with which to circumvent the issues related to T-cell aplasia. As mentioned above, several potential target antigens have been investigated, including TRBC1/2, CDR3, CD1a, CD4, CCR7, CCR9, CD30, CD33, and CD99 [9][17][18][19][21][22][43][44]. CD1a is expressed in cortical T-ALL, which comprises 35–40% of all T-ALL cases [26] and has no expression on T cells or CD34+ hematopoietic progenitors. In a preclinical study, CD1a CAR T cells have been shown to be fratricide resistant and a safe treatment for relapsed/refractory coT-ALL patients [17][45]. CD30 is expressed by a subset of activated T cells, including almost all Hodgkin lymphomas (HL) and anaplastic large cell lymphomas (ALCL), and approximately 38% of T-ALL. Furthermore, Zheng et al. reported the expression of CD3 to be upregulated during high-dose chemotherapy in T-ALL patients [46]. Currently, 11 clinical trials concerning anti-CD30 CAR T therapy in CD30-positive malignancies are ongoing (NCT01316146, NCT01192464, NCT03049449, NCT02690545, NCT02958410, NCT02663297, NCT03383965, NCT02917083, NCT04008394, NCT02259556, and NCT03602157) and each is at a different stage of clinical trials. One of the clinical trials, involving 41 patients showed an overall response rate of 72%, a CR rate of 59%, and none of the patients suffered life-threatening infections [47].
Another promising approach is the incorporation of suicide genes and switches into the CAR constructs to restrict the life and activities of CAR T cells. Gina Ma et al. used CAMPATH (alemtuzumab) as a natural safety switch to deplete the infused CD4 CAR T cells. This was shown to avert CAR T-caused CD4 cell aplasia after eliminating CD4-positive T-ALL in a systemic mouse model [9]. Their results demonstrated that CD4 CAR T cells have robust antitumor activities in vivo, and CAR T cells were depleted 24 h after CAMPATH treatment in different organs. In addition, bridging to an allogeneic HSCT following CAR T-cell therapy and using short-lived effector cells instead of T cells have been shown to be valid methods to prevent T-cell aplasia, with both strategies succeeding in clinical studies [48][49].

3. Product Contamination and Potential Strategies

One important issue in the field of CAR T-cell therapy is the contamination of CAR T-cell products with tumor cells, which subsequently generates CAR-modified tumor cells with immunity to CAR T cells [42]. The CAR molecule ‘masks’ the antigen on the tumor cells, preventing it from being recognized by CAR T cells, which has serious implications for manufacturing safety [50]. In 2018, Ruella et al. reported a patient relapsing after anti-CD19 CAR T-cell infusion with CD19 positive B-ALL that abnormally expressed the anti-CD19 CAR. CAR transduced B-cell leukemia (CARB) cells expanded out of control and the patient ultimately died of complications related to progressive leukemia [51].
Product contamination occurs when malignant and normal T cells are both isolated in the course of leukapheresis, especially for T-cell lymphoma or T-ALL [42]. In addition, the removal of cancer cells before gene transfer using cell purification methods can be used to address this issue. However, traditional cell purification methods such as fluorescence-activated cell separation and magnetic-activated cell separation have proven to be unsuitable for high-purity T-cell purification [52][53]. Recently, one group developed a label-free high-throughput cell purification method (dielectrophoretic cell purification) that was shown to achieve 100% purity with a high cell viability (greater than 90%). This indicates that this method is a feasible program for allogeneic CAR T therapy. However, circulating tumor T cells were often identified in the peripheral blood of T-ALL patients, which significantly increases the risk of transducing malignant cells when isolating autologous T cells [54][55][56].
Generating a CAR T-cell product using cells from healthy donors could be used to entirely avoid this problem and has been tested both in preclinical studies and clinical trials. In addition, the risk of GvHD and host-mediated rejection should also be avoided in allogeneic CAR T-cell therapies. In 2018, Cooper et al. demonstrated “off-the-shelf” CD7 CAR T, which deleted both CD7 and T-cell receptor alpha chain (TRAC) expression using CRISPR/Cas9. This demonstrated efficacy against T-ALL without developing xenogeneic GvHD in vivo [44][57].
Independent antigenic presentation by MHC molecules for recognition makes γδT cells ideal candidates for allogeneic cell therapy [58][59]. Currently, preclinical studies and early-phase clinical trials have demonstrated that ex vivo expanded Vγ9Vδ2 T cells display significant antitumor activities [60][61]. Laurence JN Cooper’s group demonstrated that γδCAR T cells displayed enhanced cytotoxicity against tumor cells in vitro [62]. Furthermore, Rozenbaum et al. reported the high transduction efficacy of γδ T cells compared with standard CAR T cells (60.5 ± 13.2 and 65.3 ± 18.3%, respectively) and demonstrated that γδCAR T cells targeting CD19 could be effective against CD19+ cell lines in vitro and in vivo and could also kill CD19 antigen negative leukemia cells [63]. Since γδT cells can be safely employed in the allogeneic setting and exhibit a robust antitumor response, γδT cells may represent a better candidate for universal CAR (UCAR) T therapy than αβT.

UCAR T technology is constantly evolving and can be used to prevent adverse effects by combining multiple gene editing strategies. It should be considered as a potential therapy for clinical trials.

In addition to the other three shortcomings, CAR T-cell dysfunction is also a disadvantage of CAR T therapy. It involves T-cell exhaustion (manifested as the loss of one of the following properties: expansion, secretion of effector cytokines and lysing target cells, surviving after antigen stimulation, and performing secondary antigen challenge) and T-cell senescence, meaning that they are not able to proliferate in response to antigen stimulation [64]. Costimulatory molecules, such as OX40 and 4-1BB, may be used to enhance T-cell antitumor activity [65]. Checkpoint blockade therapy has been approved for clinical use to combat T-cell exhaustion by increasing proliferation of less differentiated T-cell populations and generating a large pool of terminally exhausted T cells, which allows for therapeutic combinations with activated costimulatory molecules and produces noteworthy effects [66][67]

References

  1. Rabinowich, H.; Pricop, L.; Herberman, R.B.; Whiteside, T.L. Expression and function of CD7 molecule on human natural killer cells. J. Immunol. 1994, 152, 517–526.
  2. Chen, K.H.; Wada, M.; Pinz, K.G.; Liu, H.; Lin, K.W.; Jares, A.; Firor, A.E.; Shuai, X.; Salman, H.; Golightly, M. Preclinical targeting of aggressive T-cell malignancies using anti-CD5 chimeric antigen receptor. Leukemia 2017, 31, 2151–2160.
  3. Dalloul, A. CD5: A safeguard against autoimmunity and a shield for cancer cells. Autoimmun. Rev. 2009, 8, 349–353.
  4. Png, Y.T.; Vinanica, N.; Kamiya, T.; Shimasaki, N.; Coustan-Smith, E.; Campana, D. Blockade of CD7 expression in T cells for effective chimeric antigen receptor targeting of T-cell malignancies. Blood Adv. 2017, 1, 2348–2360.
  5. You, F.; Wang, Y.; Jiang, L.; Zhu, X.; Chen, D.; Yuan, L.; An, G.; Meng, H.; Yang, L. A novel CD7 chimeric antigen receptor-modified NK-92MI cell line targeting T-cell acute lymphoblastic leukemia. Am. J. Cancer Res. 2019, 9, 64–78.
  6. Raikar, S.S.; Fleischer, L.C.; Moot, R.; Fedanov, A.; Paik, N.Y.; Knight, K.A.; Doering, C.B.; Spencer, H.T. Development of chimeric antigen receptors targeting T-cell malignancies using two structurally different anti-CD5 antigen binding domains in NK and CRISPR-edited T cell lines. Oncoimmunology 2017, 7, e1407898.
  7. Wang, W.; Jiang, J.; Wu, C. CAR-NK for tumor immunotherapy: Clinical transformation and future prospects. Cancer Lett. 2019, 472, 175–180.
  8. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. Ebiomedicine 2020, 59, 102975.
  9. Huang, J.; Alexey, S.; Li, J.; Jones, T.; Grande, G.; Douthit, L.; Xie, J.; Chen, D.; Wu, X.; Michael, M. Unique CDR3 epitope targeting by CAR-T cells is a viable approach for treating T-cell malignancies. Leukemia 2019, 33, 2315–2319.
  10. Chen, K.H.; Wada, M.; Firor, A.; Pinz, K.; Jares, A.; Liu, H.; Salman, H.; Golightly, M.; Lan, F.; Jiang, X. Novel anti-CD3 chimeric antigen receptor targeting of aggressive T cell malignancies. Oncotarget 2016, 7, 56219–56232.
  11. Voynova, E.; Hawk, N.; Flomerfelt, F.A.; Telford, W.G.; Gress, R.E.; Kanakry, J.A.; Kovalovsky, D. Increased Activity of a NK-Specific CAR-NK Framework Targeting CD3 and CD5 for T-Cell Leukemias. Cancers 2022, 14, 524.
  12. Xu, Y.; Liu, Q.; Zhong, M.; Wang, Z.; Chen, Z.; Zhang, Y.; Xing, H.; Tian, Z.; Tang, K.; Liao, X. 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. J. Hematol. Oncol. 2019, 12, 49.
  13. Gomes-Silva, D.; Srinivasan, M.; Sharma, S.; Lee, C.M.; Wagner, D.L.; Davis, T.H.; Rouce, R.H.; Bao, G.; Brenner, M.K.; Mamonkin, M. CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies. Blood 2017, 130, 285–296.
  14. Montagner, I.; Penna, A.; Fracasso, G.; Carpanese, D.; Dalla Pietà, A.; Barbieri, V.; Zuccolotto, G. Anti-PSMA CAR-Engineered NK-92 Cells: An off-the-Shelf Cell Therapy for Prostate Cancer. Cells 2020, 9, 1382.
  15. Pinz, K.; Yakaboski, E.; Jares, A.; Liu, H.; Firor, A.E.; Chen, K.H.; Wada, M.; Salman, H.; Tse, W.; Hagag, N. Targeting T-cell malignancies using anti-CD4 CAR NK-92 cells. Oncotarget 2017, 8, 112783–112796.
  16. Kweon, S.; Phan, M.T.T.; Chun, S.; Yu, H.; Kim, J.; Kim, S.; Lee, J.; Ali, A.K.; Lee, S.H.; Kim, S.K. Expansion of Human NK Cells Using K562 Cells Expressing OX40 Ligand and Short Exposure to IL-21. Front. Immunol. 2019, 10, 879.
  17. Maciocia, P.M.; Pule, M.A. Anti-CD1a CAR T cells to selectively target T-ALL. Blood 2019, 133, 2246–2247.
  18. Maciocia, P.M.; Wawrzyniecka, P.A.; Maciocia, N.C.; Burley, A.; Karpanasamy, T.; Devereaux, S.; Hoekx, M.; O’Connor, D.; Leon, T.; Rapoz-D’Silva, T. Anti-CCR9 chimeric antigen receptor T cells for T-cell acute lymphoblastic leukemia. Blood 2022, 140, 25–37.
  19. Shi, J.; Zhang, Z.; Cen, H.; Wu, H.; Zhang, S.; Liu, J.; Leng, Y.; Ren, A.; Liu, X.; Zhang, Z. CAR T cells targeting CD99 as an approach to eradicate T-cell acute lymphoblastic leukemia without normal blood cells toxicity. J. Hematol. Oncol. 2021, 14, 162.
  20. Srivastava, S.; Riddell, S.R. Chimeric Antigen Receptor T Cell Therapy: Challenges to Bench-to-Bedside Efficacy. J. Immunol. 2018, 200, 459–468.
  21. Ferrari, M.; Baldan, V.; Ghongane, P.; Nicholson, A.; Bughda, R.; Akbar, Z.; Wawrzyniecka, P.; Maciocia, P.; Cordoba, S.; Thomas, S. Abstract 2183: Targeting TRBC1 and 2 for the treatment of T cell lymphomas. Immunology 2020, 80, 2183.
  22. Kim, M.Y.; Yu, K.R.; Kenderian, S.S.; Ruella, M.; Chen, S.; Shin, T.H.; Aljanahi, A.A.; Schreeder, D.; Klichinsky, M.; Shestova, O. Genetic Inactivation of CD33 in Hematopoietic Stem Cells to Enable CAR T Cell Immunotherapy for Acute Myeloid Leukemia. Cell 2018, 173, 1439–1453.e19.
  23. Pinz, K.; Liu, H.; Golightly, M.; Jares, A.; Lan, F.; Zieve, G.W.; Hagag, N.; Schuster, M.; Firor, A.E.; Jiang, X. Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells. Leukemia 2015, 30, 701–707.
  24. Niehues, T.; Kapaun, P.; Harms, D.O.; Burdach, S.; Kramm, C.; Körholz, D.; Janka-Schaub, G.; Göbel, U. A classification based on T cell selection-related phenotypes identifies a subgroup of childhood T-ALL with favorable outcome in the COALL studies. Leukemia 1999, 13, 614–617.
  25. Maciocia, P.M.; Wawrzyniecka, P.A.; Philip, B.; Ricciardelli, I.; Akarca, A.U.; Onuoha, S.C.; Legut, M.; Cole, D.K.; Sewell, A.K.; Gritti, G. Targeting the T cell receptor β-chain constant region for immunotherapy of T cell malignancies. Nat. Med. 2017, 23, 1416–1423.
  26. Laszlo, G.S.; Estey, E.H.; Walter, R.B. The past and future of CD33 as therapeutic target in acute myeloid leukemia. Blood Rev. 2014, 28, 143–153.
  27. Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Aikawa, V.; Morrissette, J.J.D.; Scholler, J.; Song, D.; Porter, D.L.; Carroll, M. CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia. Leukemia 2015, 29, 1637–1647.
  28. Khogeer, H.; Rahman, H.; Jain, N.; Angelova, E.A.; Yang, H.; Quesada, A.; Ok, C.Y.; Sui, D.; Wei, P.; Al Fattani, A. Early T precursor acute lymphoblastic leukaemia/lymphoma shows differential immunophenotypic characteristics including frequent CD33 expression and in vitro response to targeted CD33 therapy. Br. J. Haematol. 2019, 186, 538–548.
  29. Bond, J.; Graux, C.; Lhermitte, L.; Lara, D.; Cluzeau, T.; Leguay, T.; Cieslak, A.; Trinquand, A.; Pastoret, C.; Belhocine, M. Early Response–Based Therapy Stratification Improves Survival in Adult Early Thymic Precursor Acute Lymphoblastic Leukemia: A Group for Research on Adult Acute Lymphoblastic Leukemia Study. J. Clin. Oncol. 2017, 35, 2683–2691.
  30. Guo, R.J.; Bahmanyar, M.; Minden, M.D.; Chang, H. CD33, not early precursor T-cell phenotype, is associated with adverse outcome in adult T-cell acute lymphoblastic leukaemia. Br. J. Haematol. 2016, 172, 823–825.
  31. Maciocia, P.M.; Wawrzyniecka, P.; Maciocia, N.C.; Burley, A.; O’Connor, D.; Leon, T.E.; Rapoz-D’Silva, T.; Karpanasamy, T.; Pule, M.; Mansour, M.R. Anti-CCR9 CAR-T Cells for T Acute Lymphoblastic Leukemia. Blood 2021, 138 (Suppl. S1), 903.
  32. Maciocia, N.C.; Burley, A.; Nannini, F.; Wawrzyniecka, P.; Neves, M.; Karpanasamy, T.; Ferrari, M.; Marafioti, T.; Onuoha, S.; Khwaja, A. Anti-CD21 Chimeric Antigen Receptor (CAR)-T Cells for T Cell Acute Lymphoblastic Leukaemia (T-ALL). Blood 2021, 138 (Suppl. S1), 902.
  33. Rasaiyaah, J.; Georgiadis, C.; Preece, R.; Mock, U.; Qasim, W. TCRαβ/CD3 disruption enables CD3-specific antileukemic T cell immunotherapy. JCI Insight 2018, 3, e99442.
  34. Liu, X.; Zhang, Y.; Cheng, C.; Cheng, A.W.; Zhang, X.; Li, N.; Xia, C.; Wei, X.; Liu, X.; Wang, H. CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res. 2016, 27, 154–157.
  35. Yi, Y.; Chai, X.; Zheng, L.; Zhang, Y.; Shen, J.; Hu, B.; Tao, G. CRISPR-edited CART with GM-CSF knockout and auto secretion of IL6 and IL1 blockers in patients with hematologic malignancy. Cell Discov. 2021, 7, 27.
  36. Mollanoori, H.; Shahraki, H.; Rahmati, Y.; Teimourian, S. CRISPR/Cas9 and CAR-T cell, collaboration of two revolutionary technologies in cancer immunotherapy, an instruction for successful cancer treatment. Hum. Immunol. 2018, 79, 876–882.
  37. Mamonkin, M.; Mukherjee, M.; Srinivasan, M.; Sharma, S.; Gomes-Silva, D.; Mo, F.; Krenciute, G.; Orange, J.S.; Brenner, M.K. Reversible Transgene Expression Reduces Fratricide and Permits 4-1BB Costimulation of CAR T Cells Directed to T-cell Malignancies. Cancer Immunol. Res. 2017, 6, 47–58.
  38. Diorio, C.; Murray, R.; Naniong, M.; Barrera, L.; Camblin, A.; Chukinas, J.; Coholan, L.; Edwards, A.; Fuller, T.; Gonzales, C. Cytosine base editing enables quadruple-edited allogeneic CART cells for T-ALL. Blood 2022, 140, 619–629.
  39. Zhang, M.; Fu, X.; Meng, H.; Wang, M.; Wang, Y.; Zhang, L.; Li, L.; Li, X.; Wang, X.; Sun, Z. A Single-Arm, Open-Label, Pilot Trial of Autologous CD7-CAR-T Cells for CD7 Positive Relapsed and Refractory T-Lymphoblastic Leukemia/Lymphoma. Blood 2021, 138, 3829.
  40. Pan, J.; Tan, Y.; Wang, G.; Deng, B.; Ling, Z.; Song, W.; Seery, S.; Zhang, Y.; Peng, S.; Xu, J. Donor-Derived CD7 Chimeric Antigen Receptor T Cells for T-Cell Acute Lymphoblastic Leukemia: First-in-Human, Phase I Trial. J. Clin. Oncol. 2021, 39, 3340–3351.
  41. Daniël, K.; Gäken, J.; Farzaneh, F.; Kemeny, D.M. The Use of Intracellular Single-Chain Antibody Fragments to Specifically Inhibit Cytokine Secretion. Int. Arch. Allergy Immunol. 2001, 124, 216–217.
  42. Alcantara, M.; Tesio, M.; June, C.H.; Houot, R. CAR T-cells for T-cell malignancies: Challenges in distinguishing between therapeutic, normal, and neoplastic T-cells. Leukemia 2018, 32, 2307–2315.
  43. Ma, G.; Shen, J.; Pinz, K.; Wada, M.; Park, J.; Kim, S.; Togano, T.; Tse, W. Targeting T Cell Malignancies Using CD4CAR T-Cells and Implementing a Natural Safety Switch. Stem Cell Rev. Rep. 2019, 15, 443–447.
  44. Boyiadzis, M.M.; Dhodapkar, M.V.; Brentjens, R.J.; Kochenderfer, J.N.; Neelapu, S.S.; Maus, M.V.; Porter, D.L.; Maloney, D.G.; Grupp, S.A.; Mackall, C.L. Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: Clinical perspective and significance. J. Immunother. Cancer 2018, 6, 137.
  45. Sánchez-Martínez, D.; Baroni, M.L.; Gutierrez-Agüera, F.; Roca-Ho, H.; Blanch-Lombarte, O.; González-García, S.; Torrebadell, M.; Junca, J.; Ramírez-Orellana, M.A.; Velasco-Hernández, T.A.; et al. Fratricide-resistant CD1a-specific CAR T cells for the treatment of cortical T-cell acute lymphoblastic leukemia. Blood 2019, 133, 2291–2304.
  46. Zheng, W.; Medeiros, L.J.; Young, K.H.; Goswami, M.; Powers, L.; Kantarjian, H.H.; Thomas, D.A.; Cortes, J.E.; Wang, S.A. CD30 expression in acute lymphoblastic leukemia as assessed by flow cytometry analysis. Leuk. Lymphoma 2013, 55, 624–627.
  47. Ramos, C.A.; Grover, N.S.; Beaven, A.W.; Lulla, P.D.; Wu, M.F.; Ivanova, A.; Wang, T.; Shea, T.C.; Rooney, C.M.; Dittus, C. Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin Lymphoma. J. Clin. Oncol. 2020, 38, 3794–3804.
  48. Jacoby, E. The role of allogeneic HSCT after CAR T cells for acute lymphoblastic leukemia. Bone Marrow Transplant. 2019, 54, 810–814.
  49. Hotblack, A.; Kokalaki, E.K.; Palton, M.J.; Cheung, G.W.K.; Williams, I.P.; Manzoor, S.; Grothier, T.I.; Piapi, A.; Fiaccadori, V.; Wawrzyniecka, P. Tunable control of CAR T cell activity through tetracycline mediated disruption of protein–protein interaction. Sci. Rep. 2021, 11, 21902.
  50. Larson, R.C.; Maus, M.V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 2021, 21, 145–161.
  51. Ruella, M.; Xu, J.; Barrett, D.M.; Fraietta, J.A.; Reich, T.J.; Ambrose, D.E.; Klichinsky, M.; Shestova, O.; Patel, P.R.; Kulikovskaya, I. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nat. Med. 2018, 24, 1499–1503.
  52. Takahashi, N.; Miura, I.; Saitoh, K.; Miura, A.B. Lineage Involvement of Stem Cells Bearing the Philadelphia Chromosome in Chronic Myeloid Leukemia in the Chronic Phase as Shown by a Combination of Fluorescence-Activated Cell Sorting and Fluorescence In Situ Hybridization. Blood 1998, 92, 4758–4763.
  53. Handgretinger, R.; Lang, P.; Schumm, M.; Taylor, G.; Neu, S.; Koscielnak, E.; Niethammer, D.; Klingebiel, T. Isolation and transplantation of autologous peripheral CD34+ progenitor cells highly purified by magnetic-activated cell sorting. Bone Marrow Transplant. 1998, 21, 987–993.
  54. Marks, D.I.; Paietta, E.M.; Moorman, A.V.; Richards, S.M.; Buck, G.; DeWald, G.; Ferrando, A.; Fielding, A.K.; Goldstone, A.H.; Ketterling, R.P. T-cell acute lymphoblastic leukemia in adults: Clinical features, immunophenotype, cytogenetics, and outcome from the large randomized prospective trial (UKALL XII/ECOG 2993). Blood 2009, 114, 5136–5145.
  55. Litzow, M.R.; Ferrando, A.A. How I treat T-cell acute lymphoblastic leukemia in adults. Blood 2015, 126, 833–841.
  56. Longo, D.; Hunger, S.; Mullighan, C. Acute Lymphoblastic Leukemia in Children. N. Engl. J. Med. 2015, 373, 1541–1552.
  57. Cooper, M.L.; Choi, J.; Staser, K.; Ritchey, J.K.; Devenport, J.M.; Eckardt, K.; Rettig, M.P.; Wang, B.; Eissenberg, L.G.; Ghobadi, A. An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia 2018, 32, 1970–1983.
  58. Legut, M.; Cole, D.K.; Sewell, A.K. The promise of γδ T cells and the γδ T cell receptor for cancer immunotherapy. Cell. Mol. Immunol. 2015, 12, 656–668.
  59. Silva-Santos, B.; Serre, K.; Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691.
  60. Harrer, D.C.; Simon, B.; Fujii, S.I.; Shimizu, K.; Uslu, U.; Schuler, G.; Gerer, K.F.; Hoyer, S.; Dörrie, J.; Schaft, N. RNA-transfection of γ/δ T cells with a chimeric antigen receptor or an α/β T-cell receptor: A safer alternative to genetically engineered α/β T cells for the immunotherapy of melanoma. BMC Cancer 2017, 17, 551.
  61. Capsomidis, A.; Benthall, G.; Van Acker, H.H.; Fisher, J.; Kramer, A.M.; Abeln, Z.; Majani, Y.; Gileadi, T.; Wallace, R.; Gustafsson, K. Chimeric Antigen Receptor-Engineered Human Gamma Delta T Cells: Enhanced Cytotoxicity with Retention of Cross Presentation. Mol. Ther. 2018, 26, 354–365.
  62. Deniger, D.C.; Switzer, K.; Mi, T.; Maiti, S.; Hurton, L.; Singh, H.; Huls, H.; Olivares, S.; Lee, D.A.; Champlin, R.E. Bispecific T-cells Expressing Polyclonal Repertoire of Endogenous δ T-cell Receptors and Introduced CD19-specific Chimeric Antigen Receptor. Mol. Ther. J. Am. Soc. Gene Ther. 2013, 21, 638–647.
  63. Rozenbaum, M.; Meir, A.; Aharony, Y.; Itzhaki, O.; Schachter, J.; Bank, I.; Jacoby, E.; Besser, M.J. Gamma-Delta CAR-T Cells Show CAR-Directed and Independent Activity Against Leukemia. Front. Immunol. 2020, 11, 1347.
  64. Titov, A.; Kaminskiy, Y.; Ganeeva, I.; Zmievskaya, E.; Valiullina, A.; Rakhmatullina, A.; Petukhov, A.; Miftakhova, R.; Rizvanov, A.; Bulatov, E. Knowns and Unknowns about CAR-T Cell Dysfunction. Cancers 2022, 14, 1078.
  65. Taraban, V.Y.; Rowley, T.F.; O’Brien, L.; Chan, H.T.C.; Haswell, L.E.; Green, M.H.A.; Tutt, A.L.; Glennie, M.J.; Al-Shamkhani, A. Expression and costimulatory effects of the TNF receptor superfamily members CD134 (OX40) and CD137 (4–1BB), and their role in the generation of anti-tumor immune responses. Eur. J. Immunol. 2002, 32, 3617–3627.
  66. Curran, M.A.; Kim, M.; Montalvo, W.; Al-Shamkhani, A.; Allison, J.P. Combination CTLA-4 blockade and 4–1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS ONE 2011, 6, e19499.
  67. Guo, Z.; Wang, X.; Cheng, D.; Xia, Z.; Luan, M.; Zhang, S. PD-1 blockade and OX40 triggering synergistically protects against tumor growth in a murine model of ovarian cancer. PLoS ONE 2014, 9, e89350.
More
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 492
Revisions: 3 times (View History)
Update Date: 21 Apr 2023
1000/1000
Video Production Service