Next Article in Journal
Circulating miR-499a and miR-125b as Potential Predictors of Left Ventricular Ejection Fraction Improvement after Cardiac Resynchronization Therapy
Next Article in Special Issue
Induced CAR-Macrophages as a Novel Therapeutic Cell Type for Cancer Immune Cell Therapies
Previous Article in Journal
Large-Scale, Wavelet-Based Analysis of Lysosomal Trajectories and Co-Movements of Lysosomes with Nanoparticle Cargos
Previous Article in Special Issue
Transfer of Cellular Content from the Allogeneic Cell-Based Cancer Vaccine DCP-001 to Host Dendritic Cells Hinges on Phosphatidylserine and Is Enhanced by CD47 Blockade
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using iPSC Technology and Gene Editing

1
Department of Hematology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
2
Division of Cell Therapy & Blood Transfusion Medicine, Juntendo University School of Medicine, Tokyo 113-8421, Japan
3
Division of Stem Cell Therapy, Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
4
Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5461, USA
*
Authors to whom correspondence should be addressed.
Co-first authorship.
Cells 2022, 11(2), 269; https://doi.org/10.3390/cells11020269
Submission received: 8 December 2021 / Revised: 7 January 2022 / Accepted: 10 January 2022 / Published: 13 January 2022
(This article belongs to the Special Issue Allogeneic Cell Cancer Immunotherapies)

Abstract

:
The concept of allogeneic cell therapy was first presented over 60 years ago with hematopoietic stem cell transplantation. However, complications such as graft versus host disease (GVHD) and regimen-related toxicities remained as major obstacles. To maximize the effect of graft versus leukemia, while minimizing the effect of GVHD, donor lymphocyte infusion was utilized. This idea, which was used against viral infections, postulated that adoptive transfer of virus-specific cytotoxic T lymphocytes could reconstitute specific immunity and eliminate virus infected cells and led to the idea of banking third party cytotoxic T cells (CTLs). T cell exhaustion sometimes became a problem and difficulty arose in creating robust CTLs. However, the introduction of induced pluripotent stem cells (iPSCs) lessens such problems, and by using iPSC technology, unlimited numbers of allogeneic rejuvenated CTLs with robust and proliferative cytotoxic activity can be created. Despite this revolutionary concept, several concerns still exist, such as immunorejection by recipient cells and safety issues of gene editing. In this review, we describe approaches to a feasible “off-the-shelf” therapy that can be distributed rapidly worldwide. We also offer perspectives on the future of allogeneic cell cancer immunotherapy.

1. Introduction

More than 60 years ago, in 1957, Dr. E. Donnall Thomas initially reported an innovative approach towards leukemia using radiation and chemotherapy followed by intravenous infusion of bone marrow [1,2], the first report of allogeneic hematopoietic stem cell transplantation (allo-HSCT), in which a donor’s hematopoietic stem cells and his or her immunological repertoires are infused into a patient to establish donor-derived hematopoiesis and immunity [3]. Thomas et al. conducted many laboratory and clinical investigations [4,5,6,7,8,9] that paved the way for current allo-HSCT, for which, in 1990, along with Dr. Joseph E. Murray, Thomas was honored with the Nobel Prize in Physiology or Medicine. Allogeneic cell therapy has continued to improve and has endured as an effective treatment option for both malignant and non-malignant diseases, especially for hematologic malignancies such as leukemia, which was formerly thought to be an incurable disease. Although allo-HSCT is a revolutionary treatment strategy, significant complications remain; most importantly, graft-versus-host disease (GVHD) and conditioning related toxicities.
The first report of GVHD was in 1959, when Billingham and colleagues described an immune reaction after bone marrow infusion, including skin rash and diarrhea, which they called “Runt Disease” [10]. GVHD is a life-threatening complication that occurs when immunocompetent T cells in donated tissue recognize the recipient as foreign, resulting in activation of donor T cells. These gain cytotoxic capacity and then attack the recipient to eliminate “foreign” antigen-bearing cells [11]. Management of both acute and chronic GVHD severely challenged early transplantation research groups. In 1974, the first grading system for acute GVHD was created [12], increasing awareness of skin, gastrointestinal, and liver symptoms in transplanted patients. In early trials, GVHD occurred in almost half of patients who received methotrexate prophylaxis. Major progress occurred when methotrexate was combined with calcineurin inhibitors such as cyclosporine or tacrolimus, initially in canine models and subsequently in patients, with improved overall survival [13,14,15,16]. Effective treatment for acute GVHD with antithymocyte globulin (ATG) was also reported [17], and these synergistic drug combinations of methotrexate, ATG, cyclosporine, and tacrolimus are still used to prevent GVHD [18]. Unfortunately, among patients undergoing allo-HSCT, 30–50% have acute GVHD (grade I–IV), and 14% have severe acute GVHD (grade III–IV) [11]. Risk factors include the degree of human leukocyte antigen (HLA) mismatch, unrelated donor, a female donor for a male recipient, the use of peripheral blood stem cell grafts, and myeloablative conditioning [19,20,21,22].
Chronic GVHD also remains as a major cause of morbidity and non-relapse mortality after allo-HSCT [23,24,25,26]. Approximately 30–70% of allo-HSCT recipients surviving at least 100 days after transplant develop chronic GVHD [26]. The skin is the most commonly affected site, with manifestations that can be observed in up to 75% of chronic GVHD patients, but clinical features may affect multiple organs or body areas with varying presentation, depending on the patient [27,28]. Corticosteroids with or without calcineurin inhibitors compose first-line treatment, but the prognosis is poor if the patient is steroid-refractory [29].
Other than GVHD, severe immunodeficiency is ubiquitous after allogeneic transplantation. Cytomegalovirus, Epstein-Barr virus (EBV), polyomavirus, human herpesvirus 6, and community-acquired respiratory viruses substantially affect allo-HSCT related morbidity and mortality [3,30]. Although drug therapy to prevent infection is indispensable [13,31] and treatments for infection are advancing, fatal infections are still inevitable following allo-HSCT.
The difficulty of treating GVHD is to balance graft versus leukemia (GVL), where the key to remission lies in maximizing the effect of GVL while minimizing the effect of GVHD. GVL was first hypothesized in 1979 when Weiden et al. reported that the risk of leukemia relapse was 2.5 times lower in patients with GVHD than in those without GVHD [32].
Many strategies to reinforce GVL have been deployed, such as donor lymphocyte infusion (DLI) [33] and natural killer cells [34,35]. DLI has augmented GVL effectively, particularly in multiple myeloma [36,37] and chronic myeloid leukemia [38]. However, DLI may cause GVHD so severe as to be life-threatening [39]. DLI has also been successfully used after allo-HSCT against severe viral infections, such as those with EBV and adenovirus, where adoptive transfer of virus-specific cytotoxic T lymphocytes (VSTs) was postulated to be able to reconstitute specific immunity and to eliminate virus infected cells [40,41]. T cell immunology directed attention to cytotoxic T lymphocytes (CTLs), which play an important role in the immune system defense against viral infections and malignancies [42,43,44,45,46,47]. CTLs are triggered by the presentation of antigen peptides in the context of major histocompatibility complex (MHC) class I molecules, leading to target cell death [44,48,49,50]. The success of VSTs in treating severe viral infections led to the idea of banked third party VSTs, as an approach to rapid treatment of intractable viral infections after allo-HSCT [51,52,53,54,55]. Although CTLs protect us from viral and bacterial infections by destroying infected cells, this process requires T cell MHC allele recognition, which is known as MHC restriction.
However, the recent discovery of induced pluripotent stem cells (iPSCs) [56,57], along with gene editing using CRISPR/Cas9 technology, has cast light upon the road leading toward allogeneic cancer immunotherapy.
In this review we discuss new treatment strategies using iPSC technology for cancer immunotherapy, with suggestions on their prospects.

2. Adoptive T Cell Therapy and Tumor Microenvironment

Adoptive T cell therapy, with administration of antigen-specific CTLs expanded ex vivo, has shown success in infections caused by a variety of viruses [58,59,60,61,62] and in malignancies such as melanoma [63,64,65,66,67,68]. However, solid tumors are relatively insusceptible to T cell-mediated destruction in the tumor microenvironment, which is an obstacle when using adoptive T cell therapy against solid tumors [68,69].
Cancer cells utilize both passive and active defense mechanisms to survive [70]. A passive defense mechanism is the reduction of MHC expression or antigen processing ability to limit antigen detection by T cells or the editing of presented antigens by not expressing targeted antigens [71]. An active defense mechanism, by contrast, directly limits the cytotoxic capacity of T cells. For example, they may express Fas ligand that induces apoptosis in CTLs expressing the cognate receptor [72,73,74], or they may secrete soluble factors such as transforming growth factor beta and interleukin(IL)-10 [75,76,77,78] as well as chemokines that attract regulatory T cells which, in turn, inhibit effector T cell function [79,80]. Other elements, such as programmed death receptor-1(PD-1), also inhibit the proliferation and activation of effector T cell function through the recruitment of SHP2, which inactivates T cell receptor-mediated signaling [81,82,83,84].

3. T Cell Exhaustion

These passive and active defense mechanisms can lead to CTL exhaustion and deactivation. T cell exhaustion is a state in where T cell dysfunction arises in association with chronic infection and cancer [85,86]. It is defined by poor effector function, sustained expression of inhibitory receptors and an altered transcriptional program [85]. This prevents optimal control of infections and tumors, leading to apoptosis of exhausted cells. However, revitalizing exhausted T cells can reinvigorate immunity [86].

4. Use of iPSC Technology to Reinvigorate Immunity

iPSC technology effectively overcomes T cell exhaustion and bolsters immunity. iPSCs were created in 2006 [56,57]; iPSC technology has since been widely utilized in the medical field. 2014 saw the first clinical trial of autologous iPSC-derived cell therapy, in which iPSC-derived retinal pigment epithelial cells were used to palliate wet-type age-related macular degeneration [87,88]. In 2019, allogeneic iPSC-derived corneal epithelial cells were transplanted into a patient with corneal epithelial stem cell deficiency [89].
Recently, iPSC technology has been utilized in cancer immunotherapy [45,46,47].

5. Rejuvenation of CTLs

In 2013, a new strategy was created to solve the problem of T cell exhaustion by converting exhausted antigen-specific CTLs into iPSCs (T-iPSCs) [90]. This involved transducing two Sendai virus vectors, along with the Yamanaka four factors [56] and SV40 large T antigen, into a Nef-specific CTL clone derived from the peripheral blood of a human immunodeficiency virus-infected patient. These rejuvenated CTLs showed the same antigen specificity as the original CTLs throughout the whole process of reprogramming, redifferentiation, and expansion. They also proliferated better and had longer telomeres than the original CTLs, and they did not express exhaustion markers such as PD-1 [90]. Almost simultaneously, CTLs specific for melanoma epitope MART-1 were created. These secreted interferon γ, demonstrating that redifferentiation into functional MART-1-specific CTLs had occurred [91]. These findings paved the way for use of iPSC-derived CTLs in viral and malignant diseases.

6. Anti-Tumor Effect of iPSC-Derived CTLs Proven In Vivo

iPSC-derived rejuvenated CTLs applied in vivo, using EBV-specific CTLs, were first described in 2015; treatment with rejuvenated CTLs significantly prolonged survival of mice harboring EBV-infected lymphoma cells [92].
In 2020, iPSC-derived EBV antigen-specific CTLs directed against a highly aggressive lymphoma, extranodal natural killer (NK)/T cell lymphoma and nasal type (ENKL), showed promising results that included significantly prolonged survival, robust tumor suppressive effects, and persistence of administered cells as central memory T cells in vivo for at least 6 months, providing a sustainable effect [93]. ENKL cells infected by EBV express a small number of EBV genes, including those encoding latent membrane protein (LMP)1 and/or LMP2, used as an antigen target [58,61,94,95,96,97].

7. Rejuvenated CTLs for Solid Cancers

Rejuvenated CTLs have also shown promising results in other malignancies, including cervical cancer associated with human papilloma virus (HPV) infection [98] and renal cell carcinoma [99]. For cervical cancer, T-iPSCs established from HPV16 E6-or E7-CTLs efficiently differentiated into HPV16-specific rejuvenated CTLs; rejuvenated CTLs survived longer than original CTLs, while inducing increased tumor shrinkage and substantially prolonged survival in a mouse model in vivo [98]. For renal cell carcinomas, T-iPSC established from Wilms tumor 1-specific CTLs suppressed tumor growth in a mouse model in vivo [99]. Furthermore, T-iPSC technology has shown success against Ewing sarcoma [100]; rejuvenated CTLs directed against neoantigen encoded by the EWS/FLI1 fusion gene showed an anti-tumor effect in xenograft mouse models.

8. Rejuvenated CTLs as an Unlimited Source for “Off-the-Shelf” Therapy

iPSC-derived autologous CTLs are impracticably expensive to generate. Furthermore, their generation takes time—time that patients with rapidly progressing disease do not have. Allogeneic banking of third-party T-iPSCs using antigen specific CTL clones established from peripheral blood of healthy donors could be a solution for both problems [101]. Antigen-specific CTLs could be used to make various antigen-specific rejuvenated CTLs available whenever needed. While many trials using banked third-party peripheral blood-derived virus specific T cells have already shown success [51,52,53,54], use of such cells is limited by potential for expansion ex vivo. Rejuvenated CTLs proliferate strongly and could constitute an unlimited source of antigen-specific T-iPSCs to permit “off-the-shelf” therapy for patients around the world.
In establishing a T-iPSC third-party bank, the burdens of the HLA barrier must be overcome [102]. In order to do so, one must address, from a global perspective, how many T-iPSC lines are necessary to provide rejuvenated CTLs for patients around the world. In a multicenter trial, 32 virus-specific T cell lines were produced, and 18 were applied to 50 study patients. Specifically selected T cell lines matched one or more HLA alleles in 90% of the selected patients [51,52,53,54,55,101]. This suggests an approximate number of cell lines needed to cover a large population, and T-iPSC banking for “off-the-shelf” therapy could be patterned similarly.

9. iPSC Technology Exerting Its “Off-the-Shelf” Advantage in CART Therapy

CARs have also emerged as an innovative strategy for adoptive T cell therapy [103,104,105,106,107] free of HLA restrictions, meaning that cells expressing CARs can be used regardless of patient HLA types [108,109,110,111,112] (Figure 1A). Clinical trials of CART therapy directed against the antigen CD19 have shown nearly 90% complete remission in patients with relapsed or treatment-refractory acute lymphoblastic leukemia [113,114,115,116].
Although patient-derived CART therapy shows promising results, autologous CART cells are individually manufactured, meaning that quantity and quality of the cells administered are sometimes problematic. From blood collection to administration of adequate doses of an autologous CART product, it may take three or more weeks, too long for patients with rapidly progressing disease [70]. Autologous T cells exhausted by chronic exposure to tumor cells often migrate and proliferate poorly, making them unable to eradicate tumors [85,117,118,119,120,121]. “Off-the-shelf” CART therapy may offer promise in meeting these needs. Table 1 lists various strategies in allogeneic CART therapy. Unless using MHC-unrestricted cells when engrafting CARs, such as γδ T cells and NK cells, which are known to be less likely to trigger GVHD, genome editing is indispensable in allogeneic CART therapy. iPSCs are considered suitable for genetic modification, and the CAR transgene can be effectively introduced into iPSCs. Such iPSC-derived CART cells directed against CD19 antigen shrank CD19-expressing lymphoma cell lines in a mouse model [122,123]. The combination of CARs and rejuvenated CTLs (dual antigen rejuvenated CTLs; DRrejTs) was used against treatment refractory EBV-associated lymphoma, where a CAR was engineered to target the CD19 antigen and introduced into iPSC-derived from latent membrane protein 2-specific CTLs to generate DRrejTs. This strategy showed a robust anti-tumor effect and long persistence against human lymphoma in an immunodeficient mouse model. It may soon help to overcome several substantial problems for the treatment of refractory lymphoma patients [124] (Figure 1B).

10. Suicide Gene-Based Safeguard System

Gene-engineered T cells react strongly to tumor-associated antigens. This is desirable. A drawback, however, is that if such an antigen is expressed on healthy tissue, these T cells might attack that tissue, causing off-tumor toxicity along with cytokine storm [108,125,126]. These concerns have increased interest in introducing suicide genes into donor cells. The first suicide gene deployed was herpes simplex virus-thymidine kinase (HSV-TK) [127,128,129] (Figure 2A). A step forward has come with the introduction of inducible caspase-9 (iC9) protein, fused to human caspase-9 and to modified FK506-binding protein, which has overcome several concerns associated with HSV-TK [130,131,132,133,134,135,136,137,138] (Figure 2B) and has shown an effect against GVHD in leukemia [134,136,137]. This iC9 safeguard system has also shown effectiveness and safety in regenerative medicine [139,140,141,142,143]. In the treatment of a spinal cord injury, although transplantation of iPSC-derived neural stem/progenitor cells was a promising approach to amelioration of motor function, teratoma formation was a major concern. This was resolved by utilizing the iC9 safeguard system to ablate all transplanted iPSC-derived cells that hindered legitimate motor function [142]. CRISPR/Cas9 genome editing refined the system to introduce a second drug-inducible safeguard: knocking in an iC9 immediately downstream of NANOG allowed killing only iPSCs to prevent teratoma formation, whereas knocking an iC9 into ACTB allowed elimination of all cells [24,70]. This iC9 safeguard system has been used in preclinical trials for rejuvenated CTL therapy. These showed that iPSC-derived rejuvenated CTLs exerted a strong antitumor effect, and that introduction of the iC9 safeguard system did not disturb antigen-specific killing activity [92]. The system has also shown promise in CART therapy, where cytokine release syndrome has often been a problem [131,144]. Suicide gene systems based on iC9 are a promising facilitator of clinical use of iPSC-derived cell therapy.

11. Gene Edited iPSC-Derived T Cell Therapy

The greatest advantage of allogeneic rejuvenated CTL therapy lies in the ease of delivery from an unlimited source, once T-iPSC banking is established. If the T-iPSC bank is fully stocked, rejuvenated HLA-matched CTLs can be distributed. However, fully stocking the T-iPSC banking may be difficult if serving populations in which HLA haplotypes widely vary. To overcome this problem, HLA engineering has been performed in T-iPSCs from healthy donors, making it possible to create allogeneic CTLs that can be used to treat HLA-matched patients.
To overcome these complications, genome editing is essential. Many methods have been reported [54,145,146,147,148,149,150,151,152,153,154,155]. In immunocompetent patients, rejection by recipient CD8 T cells is the primary complication.
To address this, B2M, encoding β2-microglobulin (B2M), a common protein subunit necessary for surface expression of all HLA in class I, has been disrupted. However, disruption of B2M causes NK cells to attack HLA class I negative cells because they lack an essential inhibitory ligand, thus triggering the “missing-self” response [54,145,146,147,148,151]. This response was first initially reported in the setting of bone marrow transplantation where MHC-homo mouse cells were rejected by MHC-hetero recipient mice [156,157], a phenomenon eventually traced to NK cells [149,150,158]. To prevent this “missing self” response, many strategies have been assayed. B2M was knocked out to erase HLA-class I expression, and HLA-E was knocked in. Its product, HLA-E, binds to the CD94/NKG2A receptor on NK cells and protects cells that express HLA-class I from NK-dependent lysis. Thus, the overexpression of HLA-E prevents the “missing self” response [150,151]. In another approach, iPSCs overexpressed CD47, one of the “don’t eat me” signals, after the depletion of HLA molecules [146], which impaired NK cell response in B2M knock out cells. Yet another approach deleted iPSC HLA class I and II while knocking in HLA-G, CD47, and PD-L1 [152]. This strategy was supported by reports that HLA-G1 reduces the NK cell-mediated immune response [153,154,155] and by the evident fact that PD-L1 suppresses T cell activity. A different group developed HLA-homo-iPSCs in which HLA-A, HLA-B, and HLA-class II were deleted but HLA-C was retained; non-canonical HLA molecules were expected to prevent immune rejection by NK cells [148,150]. Recently, PVR knock out (PVR encodes a ligand on the NK cell-activating receptor DNAM-1), HLA-E transduced, HLA-I- and HLA-II null iPSC successfully yielded CTLs that escaped alloreactive host recognition [159] (Figure 3).
Although these techniques are very promising, they risk off-target genome editing, which might cause oncogenic events. Strategies such as single cell genome editing and introduction of a safeguard system can evade this risk. Single cell genome editing of iPSCs is considered technically feasible, and the iC9 safeguard system, as explained earlier, is also promising.

12. Conclusions

Innovative progress continues in allogeneic cell cancer immunotherapy, with advances toward clinical use. Banking of third-party T-iPSCs or generation of hypoimmunogenic iPSC by HLA editing, with the banked cells as an unlimited source of therapeutic T cells, would be a feasible approach for “off-the-shelf” therapy available for urgent use at a reasonable cost [51,52,53,54,101]. Introduction of T-iPSC technology into T cell therapy could thus confer better clinical prognosis. Incorporation of the iC9 system constitutes an efficient safeguard for iPSC-derived cell therapy. These approaches are paving the way into a new and promising era for allogeneic immunotherapy in cancer treatment.

Author Contributions

Conceptualization, H.N. and M.A.; writing—original draft preparation, Y.F.; writing—review and editing, Y.H., S.S., Y.A. and J.A.; visualization, S.K.; supervision, Y.H, J.A., H.N. and M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank A.S. Knisely for the critical reading of the manuscript.

Conflicts of Interest

H.N. is a co-founder of and an advisor to Century Therapeutics.

References

  1. Thomas, E.D.; Lochte, H.L., Jr.; Lu, W.C.; Ferrebee, J.W. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N. Engl. J. Med. 1957, 257, 491–496. [Google Scholar] [CrossRef]
  2. Appelbaum, F.R. Hematopoietic-cell transplantation at 50. N. Engl. J. Med. 2007, 357, 1472–1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Singh, A.K.; McGuirk, J.P. Allogeneic Stem Cell Transplantation: A Historical and Scientific Overview. Cancer Res. 2016, 76, 6445–6451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Thomas, E.D.; Buckner, C.D.; Banaji, M.; Clift, R.A.; Fefer, A.; Flournoy, N.; Goodell, B.W.; Hickman, R.O.; Lerner, K.G.; Neiman, P.E.; et al. One hundred patients with acute leukemia treated by chemotherapy, total body irradiation, and allogeneic marrow transplantation. Blood 1977, 49, 511–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Thomas, E.D.; Buckner, C.D.; Clift, R.A.; Fefer, A.; Johnson, F.L.; Neiman, P.E.; Sale, G.E.; Sanders, J.E.; Singer, J.W.; Shulman, H.; et al. Marrow transplantation for acute nonlymphoblastic leukemia in first remission. N. Engl. J. Med. 1979, 301, 597–599. [Google Scholar] [CrossRef]
  6. Thomas, E.D.; Buckner, C.D.; Rudolph, R.H.; Fefer, A.; Storb, R.; Neiman, P.E.; Bryant, J.I.; Chard, R.L.; Clift, R.A.; Epstein, R.B.; et al. Allogeneic marrow grafting for hematologic malignancy using HL-A matched donor-recipient sibling pairs. Blood 1971, 38, 267–287. [Google Scholar] [CrossRef]
  7. Hansen, J.A.; Clift, R.A.; Thomas, E.D.; Buckner, C.D.; Storb, R.; Giblett, E.R. Transplantation of marrow from an unrelated donor to a patient with acute leukemia. N. Engl. J. Med. 1980, 303, 565–567. [Google Scholar] [CrossRef]
  8. Tsoi, M.S.; Storb, R.; Dobbs, S.; Thomas, E.D. Specific suppressor cells in graft-host tolerance of HLA-identical marrow transplantation. Nature 1981, 292, 355–357. [Google Scholar] [CrossRef]
  9. Thomas, E.D. A history of haemopoietic cell transplantation. Br. J. Haematol. 1999, 105, 330–339. [Google Scholar] [CrossRef] [Green Version]
  10. Billingham, R.E.; Brent, L. Quantitative studies on tissue transplantation immunity IV. Induction of tolerance in newborn mice and studies on the phenomenon of runt disease. Philos. Trans. R. Soc. B 1959, 242, 477. [Google Scholar] [CrossRef]
  11. Zeiser, R.; Blazar, B.R. Acute Graft-versus-Host Disease-Biologic Process, Prevention, and Therapy. N. Engl. J. Med. 2017, 377, 2167–2179. [Google Scholar] [CrossRef]
  12. Glucksberg, H.; Storb, R.; Fefer, A.; Buckner, C.D.; Neiman, P.E.; Clift, R.A.; Lerner, K.G.; Thomas, E.D. Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors. Transplantation 1974, 18, 295–304. [Google Scholar] [CrossRef]
  13. Deeg, H.J.; Storb, R.; Weiden, P.L.; Raff, R.F.; Sale, G.E.; Atkinson, K.; Graham, T.C.; Thomas, E.D. Cyclosporin A and methotrexate in canine marrow transplantation: Engraftment, graft-versus-host disease, and induction of intolerance. Transplantation 1982, 34, 30–35. [Google Scholar] [CrossRef]
  14. Storb, R.; Deeg, H.J.; Farewell, V.; Doney, K.; Appelbaum, F.; Beatty, P.; Bensinger, W.; Buckner, C.D.; Clift, R.; Hansen, J.; et al. Marrow transplantation for severe aplastic anemia: Methotrexate alone compared with a combination of methotrexate and cyclosporine for prevention of acute graft-versus-host disease. Blood 1986, 68, 119–125. [Google Scholar] [CrossRef] [Green Version]
  15. Storb, R.; Deeg, H.J.; Whitehead, J.; Appelbaum, F.; Beatty, P.; Bensinger, W.; Buckner, C.D.; Clift, R.; Doney, K.; Farewell, V.; et al. Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft versus host disease after marrow transplantation for leukemia. N. Engl. J. Med. 1986, 314, 729–735. [Google Scholar] [CrossRef] [PubMed]
  16. Storb, R.; Raff, R.F.; Appelbaum, F.R.; Deeg, H.J.; Fitzsimmons, W.; Graham, T.C.; Pepe, M.; Pettinger, M.; Sale, G.; van der Jagt, R.; et al. FK-506 and methotrexate prevent graft-versus-host disease in dogs given 9.2 Gy total body irradiation and marrow grafts from unrelated dog leukocyte antigen-nonidentical donors. Transplantation 1993, 56, 800–807. [Google Scholar] [CrossRef] [PubMed]
  17. Storb, R.; Gluckman, E.; Thomas, E.D.; Buckner, C.D.; Clift, R.A.; Fefer, A.; Glucksberg, H.; Graham, T.C.; Johnson, F.L.; Lerner, K.G.; et al. Treatment of established human graft-versus-host disease by antithymocyte globulin. Blood 1974, 44, 56–75. [Google Scholar] [CrossRef]
  18. Granot, N.; Storb, R. History of hematopoietic cell transplantation: Challenges and progress. Haematologica 2020, 105, 2716–2729. [Google Scholar] [CrossRef] [PubMed]
  19. Riesner, K.; Shi, Y.; Jacobi, A.; Kräter, M.; Kalupa, M.; McGearey, A.; Mertlitz, S.; Cordes, S.; Schrezenmeier, J.F.; Mengwasser, J.; et al. Initiation of acute graft-versus-host disease by angiogenesis. Blood 2017, 129, 2021–2032. [Google Scholar] [CrossRef] [Green Version]
  20. Loiseau, P.; Busson, M.; Balere, M.L.; Dormoy, A.; Bignon, J.D.; Gagne, K.; Gebuhrer, L.; Dubois, V.; Jollet, I.; Bois, M.; et al. HLA Association with hematopoietic stem cell transplantation outcome: The number of mismatches at HLA-A, -B, -C, -DRB1, or -DQB1 is strongly associated with overall survival. Biol. Blood Marrow Transplant. 2007, 13, 965–974. [Google Scholar] [CrossRef] [Green Version]
  21. Flowers, M.E.; Inamoto, Y.; Carpenter, P.A.; Lee, S.J.; Kiem, H.P.; Petersdorf, E.W.; Pereira, S.E.; Nash, R.A.; Mielcarek, M.; Fero, M.L.; et al. Comparative analysis of risk factors for acute graft-versus-host disease and for chronic graft-versus-host disease according to National Institutes of Health consensus criteria. Blood 2011, 117, 3214–3219. [Google Scholar] [CrossRef] [PubMed]
  22. Jagasia, M.; Arora, M.; Flowers, M.E.; Chao, N.J.; McCarthy, P.L.; Cutler, C.S.; Urbano-Ispizua, A.; Pavletic, S.Z.; Haagenson, M.D.; Zhang, M.J.; et al. Risk factors for acute GVHD and survival after hematopoietic cell transplantation. Blood 2012, 119, 296–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Flowers, M.E.; Martin, P.J. How we treat chronic graft-versus-host disease. Blood 2015, 125, 606–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Martin, P.J.; Counts, G.W., Jr.; Appelbaum, F.R.; Lee, S.J.; Sanders, J.E.; Deeg, H.J.; Flowers, M.E.; Syrjala, K.L.; Hansen, J.A.; Storb, R.F.; et al. Life expectancy in patients surviving more than 5 years after hematopoietic cell transplantation. J. Clin. Oncol. 2010, 28, 1011–1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wingard, J.R.; Majhail, N.S.; Brazauskas, R.; Wang, Z.; Sobocinski, K.A.; Jacobsohn, D.; Sorror, M.L.; Horowitz, M.M.; Bolwell, B.; Rizzo, J.D.; et al. Long-term survival and late deaths after allogeneic hematopoietic cell transplantation. J. Clin. Oncol. 2011, 29, 2230–2239. [Google Scholar] [CrossRef] [PubMed]
  26. MacDonald, K.P.; Hill, G.R.; Blazar, B.R. Chronic graft-versus-host disease: Biological insights from preclinical and clinical studies. Blood 2017, 129, 13–21. [Google Scholar] [CrossRef]
  27. Lee, S.J.; Vogelsang, G.; Flowers, M.E. Chronic graft-versus-host disease. Biol. Blood Marrow Transplant. 2003, 9, 215–233. [Google Scholar] [CrossRef] [Green Version]
  28. Ferrara, J.L.; Levine, J.E.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef]
  29. Wolff, D.; Fatobene, G.; Rocha, V.; Kröger, N.; Flowers, M.E. Steroid-refractory chronic graft-versus-host disease: Treatment options and patient management. Bone Marrow Transplant. 2021, 56, 2079–2087. [Google Scholar] [CrossRef]
  30. Tzannou, I.; Papadopoulou, A.; Naik, S.; Leung, K.; Martinez, C.A.; Ramos, C.A.; Carrum, G.; Sasa, G.; Lulla, P.; Watanabe, A.; et al. Off-the-Shelf Virus-Specific T Cells to Treat BK Virus, Human Herpesvirus 6, Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections After Allogeneic Hematopoietic Stem-Cell Transplantation. J. Clin. Oncol. 2017, 35, 3547–3557. [Google Scholar] [CrossRef]
  31. Reisner, Y.; Kapoor, N.; Kirkpatrick, D.; Pollack, M.S.; Dupont, B.; Good, R.A.; O’Reilly, R.J. Transplantation for acute leukaemia with HLA-A and B nonidentical parental marrow cells fractionated with soybean agglutinin and sheep red blood cells. Lancet 1981, 2, 327–331. [Google Scholar] [CrossRef]
  32. Weiden, P.L.; Flournoy, N.; Thomas, E.D.; Prentice, R.; Fefer, A.; Buckner, C.D.; Storb, R. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N. Engl. J. Med. 1979, 300, 1068–1073. [Google Scholar] [CrossRef]
  33. Kolb, H.J.; Mittermüller, J.; Clemm, C.; Holler, E.; Ledderose, G.; Brehm, G.; Heim, M.; Wilmanns, W. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood 1990, 76, 2462–2465. [Google Scholar] [CrossRef] [Green Version]
  34. Yokoyama, W.M.; Plougastel, B.F. Immune functions encoded by the natural killer gene complex. Nat. Rev. Immunol. 2003, 3, 304–316. [Google Scholar] [CrossRef]
  35. Ruggeri, L.; Capanni, M.; Casucci, M.; Volpi, I.; Tosti, A.; Perruccio, K.; Urbani, E.; Negrin, R.S.; Martelli, M.F.; Velardi, A. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood 1999, 94, 333–339. [Google Scholar] [CrossRef]
  36. Lokhorst, H.M.; Schattenberg, A.; Cornelissen, J.J.; Thomas, L.L.; Verdonck, L.F. Donor leukocyte infusions are effective in relapsed multiple myeloma after allogeneic bone marrow transplantation. Blood 1997, 90, 4206–4211. [Google Scholar] [CrossRef]
  37. Kröger, N.; Badbaran, A.; Lioznov, M.; Schwarz, S.; Zeschke, S.; Hildebrand, Y.; Ayuk, F.; Atanackovic, D.; Schilling, G.; Zabelina, T.; et al. Post-transplant immunotherapy with donor-lymphocyte infusion and novel agents to upgrade partial into complete and molecular remission in allografted patients with multiple myeloma. Exp. Hematol. 2009, 37, 791–798. [Google Scholar] [CrossRef]
  38. Kolb, H.J.; Schattenberg, A.; Goldman, J.M.; Hertenstein, B.; Jacobsen, N.; Arcese, W.; Ljungman, P.; Ferrant, A.; Verdonck, L.; Niederwieser, D.; et al. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood 1995, 86, 2041–2050. [Google Scholar] [CrossRef] [Green Version]
  39. Donato, M.L.; Siegel, D.S.; Vesole, D.H.; McKiernan, P.; Nyirenda, T.; Pecora, A.L.; Baker, M.; Goldberg, S.L.; Mato, A.; Goy, A.; et al. The graft-versus-myeloma effect: Chronic graft-versus-host disease but not acute graft-versus-host disease prolongs survival in patients with multiple myeloma receiving allogeneic transplantation. Biol. Blood Marrow Transplant. 2014, 20, 1211–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Hromas, R.; Cornetta, K.; Srour, E.; Blanke, C.; Broun, E.R. Donor leukocyte infusion as therapy of life-threatening adenoviral infections after T-cell-depleted bone marrow transplantation. Blood 1994, 84, 1689–1690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Kishi, Y.; Kami, M.; Oki, Y.; Kazuyama, Y.; Kawabata, M.; Miyakoshi, S.; Morinaga, S.; Suzuki, R.; Mori, S.; Muto, Y. Donor lymphocyte infusion for treatment of life-threatening respiratory syncytial virus infection following bone marrow transplantation. Bone Marrow Transplant. 2000, 26, 573–576. [Google Scholar] [CrossRef]
  42. Greenberg, P.D. Adoptive T cell therapy of tumors: Mechanisms operative in the recognition and elimination of tumor cells. Adv. Immunol. 1991, 49, 281–355. [Google Scholar] [CrossRef]
  43. Cheever, M.A.; Chen, W. Therapy with cultured T cells: Principles revisited. Immunol. Rev. 1997, 157, 177–194. [Google Scholar] [CrossRef]
  44. Barry, M.; Bleackley, R.C. Cytotoxic T lymphocytes: All roads lead to death. Nat. Rev. Immunol. 2002, 2, 401–409. [Google Scholar] [CrossRef]
  45. Rosenberg, S.A.; Restifo, N.P.; Yang, J.C.; Morgan, R.A.; Dudley, M.E. Adoptive cell transfer: A clinical path to effective cancer immunotherapy. Nat. Rev. Cancer 2008, 8, 299–308. [Google Scholar] [CrossRef]
  46. Restifo, N.P.; Dudley, M.E.; Rosenberg, S.A. Adoptive immunotherapy for cancer: Harnessing the T cell response. Nat. Rev. Immunol. 2012, 12, 269–281. [Google Scholar] [CrossRef]
  47. Leen, A.M.; Heslop, H.E.; Brenner, M.K. Antiviral T-cell therapy. Immunol. Rev. 2014, 258, 12–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Henkart, P.A. Mechanism of lymphocyte-mediated cytotoxicity. Annu. Rev. Immunol. 1985, 3, 31–58. [Google Scholar] [CrossRef]
  49. Froelich, C.J.; Orth, K.; Turbov, J.; Seth, P.; Gottlieb, R.; Babior, B.; Shah, G.M.; Bleackley, R.C.; Dixit, V.M.; Hanna, W. New paradigm for lymphocyte granule-mediated cytotoxicity. Target cells bind and internalize granzyme B, but an endosomolytic agent is necessary for cytosolic delivery and subsequent apoptosis. J. Biol. Chem. 1996, 271, 29073–29079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Pinkoski, M.J.; Hobman, M.; Heibein, J.A.; Tomaselli, K.; Li, F.; Seth, P.; Froelich, C.J.; Bleackley, R.C. Entry and trafficking of granzyme B in target cells during granzyme B-perforin-mediated apoptosis. Blood 1998, 92, 1044–1054. [Google Scholar] [CrossRef] [PubMed]
  51. Eiz-Vesper, B.; Maecker-Kolhoff, B.; Blasczyk, R. Adoptive T-cell immunotherapy from third-party donors: Characterization of donors and set up of a T-cell donor registry. Front. Immunol. 2012, 3, 410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Barker, J.N.; Doubrovina, E.; Sauter, C.; Jaroscak, J.J.; Perales, M.A.; Doubrovin, M.; Prockop, S.E.; Koehne, G.; O’Reilly, R.J. Successful treatment of EBV-associated posttransplantation lymphoma after cord blood transplantation using third-party EBV-specific cytotoxic T lymphocytes. Blood 2010, 116, 5045–5049. [Google Scholar] [CrossRef] [PubMed]
  53. Leen, A.M.; Bollard, C.M.; Mendizabal, A.M.; Shpall, E.J.; Szabolcs, P.; Antin, J.H.; Kapoor, N.; Pai, S.Y.; Rowley, S.D.; Kebriaei, P.; et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 2013, 121, 5113–5123. [Google Scholar] [CrossRef] [PubMed]
  54. Gerdemann, U.; Katari, U.L.; Papadopoulou, A.; Keirnan, J.M.; Craddock, J.A.; Liu, H.; Martinez, C.A.; Kennedy-Nasser, A.; Leung, K.S.; Gottschalk, S.M.; et al. Safety and clinical efficacy of rapidly-generated trivirus-directed T cells as treatment for adenovirus, EBV, and CMV infections after allogeneic hematopoietic stem cell transplant. Mol. Ther. 2013, 21, 2113–2121. [Google Scholar] [CrossRef] [Green Version]
  55. Alt, F.W.; Baltimore, D. Joining of immunoglobulin heavy chain gene segments: Implications from a chromosome with evidence of three D-JH fusions. Proc. Natl. Acad. Sci. USA 1982, 79, 4118–4122. [Google Scholar] [CrossRef] [Green Version]
  56. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [Green Version]
  57. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [Green Version]
  58. Rooney, C.M.; Smith, C.A.; Ng, C.Y.; Loftin, S.; Li, C.; Krance, R.A.; Brenner, M.K.; Heslop, H.E. Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation. Lancet 1995, 345, 9–13. [Google Scholar] [CrossRef]
  59. Rooney, C.M.; Smith, C.A.; Ng, C.Y.; Loftin, S.K.; Sixbey, J.W.; Gan, Y.; Srivastava, D.K.; Bowman, L.C.; Krance, R.A.; Brenner, M.K.; et al. Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. Blood 1998, 92, 1549–1555. [Google Scholar] [CrossRef]
  60. Straathof, K.C.; Bollard, C.M.; Popat, U.; Huls, M.H.; Lopez, T.; Morriss, M.C.; Gresik, M.V.; Gee, A.P.; Russell, H.V.; Brenner, M.K.; et al. Treatment of nasopharyngeal carcinoma with Epstein-Barr virus--specific T lymphocytes. Blood 2005, 105, 1898–1904. [Google Scholar] [CrossRef] [Green Version]
  61. Comoli, P.; Pedrazzoli, P.; Maccario, R.; Basso, S.; Carminati, O.; Labirio, M.; Schiavo, R.; Secondino, S.; Frasson, C.; Perotti, C.; et al. Cell therapy of stage IV nasopharyngeal carcinoma with autologous Epstein-Barr virus-targeted cytotoxic T lymphocytes. J. Clin. Oncol. 2005, 23, 8942–8949. [Google Scholar] [CrossRef]
  62. Bollard, C.M.; Gottschalk, S.; Torrano, V.; Diouf, O.; Ku, S.; Hazrat, Y.; Carrum, G.; Ramos, C.; Fayad, L.; Shpall, E.J.; et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J. Clin. Oncol. 2014, 32, 798–808. [Google Scholar] [CrossRef]
  63. Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A.; et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N. Engl. J. Med. 1988, 319, 1676–1680. [Google Scholar] [CrossRef]
  64. Dudley, M.E.; Wunderlich, J.R.; Robbins, P.F.; Yang, J.C.; Hwu, P.; Schwartzentruber, D.J.; Topalian, S.L.; Sherry, R.; Restifo, N.P.; Hubicki, A.M.; et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002, 298, 850–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Dudley, M.E.; Wunderlich, J.R.; Yang, J.C.; Sherry, R.M.; Topalian, S.L.; Restifo, N.P.; Royal, R.E.; Kammula, U.; White, D.E.; Mavroukakis, S.A.; et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J. Clin. Oncol. 2005, 23, 2346–2357. [Google Scholar] [CrossRef] [PubMed]
  66. Rapoport, A.P.; Stadtmauer, E.A.; Aqui, N.; Badros, A.; Cotte, J.; Chrisley, L.; Veloso, E.; Zheng, Z.; Westphal, S.; Mair, R.; et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat. Med. 2005, 11, 1230–1237. [Google Scholar] [CrossRef] [PubMed]
  67. Morgan, R.A.; Chinnasamy, N.; Abate-Daga, D.; Gros, A.; Robbins, P.F.; Zheng, Z.; Dudley, M.E.; Feldman, S.A.; Yang, J.C.; Sherry, R.M.; et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 2013, 36, 133–151. [Google Scholar] [CrossRef] [Green Version]
  68. Leen, A.M.; Rooney, C.M.; Foster, A.E. Improving T cell therapy for cancer. Annu. Rev. Immunol. 2007, 25, 243–265. [Google Scholar] [CrossRef] [PubMed]
  69. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [Green Version]
  70. Ando, M.; Kinoshita, S.; Furukawa, Y.; Ando, J.; Nakauchi, H.; Brenner, M.K. Chapter 3—Improving the safety of iPSC-derived T cell therapy. In Molecular Players in iPSC Technology; Birbrair, A., Ed.; Academic Press: Cambridge, MA, USA, 2022; Volume 12, pp. 95–115. [Google Scholar]
  71. Strand, S.; Hofmann, W.J.; Hug, H.; Müller, M.; Otto, G.; Strand, D.; Mariani, S.M.; Stremmel, W.; Krammer, P.H.; Galle, P.R. Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing tumor cells--a mechanism of immune evasion? Nat. Med. 1996, 2, 1361–1366. [Google Scholar] [CrossRef] [PubMed]
  72. Hahne, M.; Rimoldi, D.; Schröter, M.; Romero, P.; Schreier, M.; French, L.E.; Schneider, P.; Bornand, T.; Fontana, A.; Lienard, D.; et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: Implications for tumor immune escape. Science 1996, 274, 1363–1366. [Google Scholar] [CrossRef] [PubMed]
  73. Bennett, M.W.; O’Connell, J.; O’Sullivan, G.C.; Brady, C.; Roche, D.; Collins, J.K.; Shanahan, F. The Fas counterattack in vivo: Apoptotic depletion of tumor-infiltrating lymphocytes associated with Fas ligand expression by human esophageal carcinoma. J. Immunol. 1998, 160, 5669–5675. [Google Scholar]
  74. Okada, K.; Komuta, K.; Hashimoto, S.; Matsuzaki, S.; Kanematsu, T.; Koji, T. Frequency of apoptosis of tumor-infiltrating lymphocytes induced by fas counterattack in human colorectal carcinoma and its correlation with prognosis. Clin. Cancer Res. 2000, 6, 3560–3564. [Google Scholar] [PubMed]
  75. Massagué, J.; Blain, S.W.; Lo, R.S. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 2000, 103, 295–309. [Google Scholar] [CrossRef] [Green Version]
  76. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.K.; Flavell, R.A. Transforming growth factor-beta regulation of immune responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
  77. Peng, Y.; Laouar, Y.; Li, M.O.; Green, E.A.; Flavell, R.A. TGF-beta regulates in vivo expansion of Foxp3-expressing CD4+CD25+ regulatory T cells responsible for protection against diabetes. Proc. Natl. Acad. Sci. USA 2004, 101, 4572–4577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Karimi, S.; Chattopadhyay, S.; Chakraborty, N.G. Manipulation of regulatory T cells and antigen-specific cytotoxic T lymphocyte-based tumour immunotherapy. Immunology 2015, 144, 186–196. [Google Scholar] [CrossRef] [Green Version]
  79. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  80. Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef] [Green Version]
  81. Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [Green Version]
  82. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [Google Scholar] [CrossRef] [PubMed]
  83. Chemnitz, J.M.; Parry, R.V.; Nichols, K.E.; June, C.H.; Riley, J.L. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J. Immunol. 2004, 173, 945–954. [Google Scholar] [CrossRef]
  84. Okazaki, T.; Maeda, A.; Nishimura, H.; Kurosaki, T.; Honjo, T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc. Natl. Acad. Sci. USA 2001, 98, 13866–13871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef]
  86. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef] [PubMed]
  87. Scudellari, M. How iPS cells changed the world. Nature 2016, 534, 310–312. [Google Scholar] [CrossRef]
  88. Kimbrel, E.A.; Lanza, R. Current status of pluripotent stem cells: Moving the first therapies to the clinic. Nat. Rev. Drug Discov. 2015, 14, 681–692. [Google Scholar] [CrossRef]
  89. Hayashi, R.; Ishikawa, Y.; Ito, M.; Kageyama, T.; Takashiba, K.; Fujioka, T.; Tsujikawa, M.; Miyoshi, H.; Yamato, M.; Nakamura, Y.; et al. Generation of corneal epithelial cells from induced pluripotent stem cells derived from human dermal fibroblast and corneal limbal epithelium. PLoS ONE 2012, 7, e45435. [Google Scholar] [CrossRef] [Green Version]
  90. Nishimura, T.; Kaneko, S.; Kawana-Tachikawa, A.; Tajima, Y.; Goto, H.; Zhu, D.; Nakayama-Hosoya, K.; Iriguchi, S.; Uemura, Y.; Shimizu, T.; et al. Generation of rejuvenated antigen-specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell 2013, 12, 114–126. [Google Scholar] [CrossRef] [Green Version]
  91. Vizcardo, R.; Masuda, K.; Yamada, D.; Ikawa, T.; Shimizu, K.; Fujii, S.; Koseki, H.; Kawamoto, H. Regeneration of human tumor antigen-specific T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell 2013, 12, 31–36. [Google Scholar] [CrossRef] [Green Version]
  92. Ando, M.; Nishimura, T.; Yamazaki, S.; Yamaguchi, T.; Kawana-Tachikawa, A.; Hayama, T.; Nakauchi, Y.; Ando, J.; Ota, Y.; Takahashi, S.; et al. A Safeguard System for Induced Pluripotent Stem Cell-Derived Rejuvenated T Cell Therapy. Stem Cell Rep. 2015, 5, 597–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ando, M.; Ando, J.; Yamazaki, S.; Ishii, M.; Sakiyama, Y.; Harada, S.; Honda, T.; Yamaguchi, T.; Nojima, M.; Ohshima, K.; et al. Long-term eradication of extranodal natural killer/T-cell lymphoma, nasal type, by induced pluripotent stem cell-derived Epstein-Barr virus-specific rejuvenated T cells in vivo. Haematologica 2020, 105, 796–807. [Google Scholar] [CrossRef] [Green Version]
  94. Gottschalk, S.; Edwards, O.L.; Sili, U.; Huls, M.H.; Goltsova, T.; Davis, A.R.; Heslop, H.E.; Rooney, C.M. Generating CTLs against the subdominant Epstein-Barr virus LMP1 antigen for the adoptive immunotherapy of EBV-associated malignancies. Blood 2003, 101, 1905–1912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Heslop, H.E.; Slobod, K.S.; Pule, M.A.; Hale, G.A.; Rousseau, A.; Smith, C.A.; Bollard, C.M.; Liu, H.; Wu, M.F.; Rochester, R.J.; et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 2010, 115, 925–935. [Google Scholar] [CrossRef] [Green Version]
  96. Bollard, C.M.; Gottschalk, S.; Leen, A.M.; Weiss, H.; Straathof, K.C.; Carrum, G.; Khalil, M.; Wu, M.F.; Huls, M.H.; Chang, C.C.; et al. Complete responses of relapsed lymphoma following genetic modification of tumor-antigen presenting cells and T-lymphocyte transfer. Blood 2007, 110, 2838–2845. [Google Scholar] [CrossRef] [PubMed]
  97. Bollard, C.M.; Rooney, C.M.; Heslop, H.E. T-cell therapy in the treatment of post-transplant lymphoproliferative disease. Nat. Rev. Clin. Oncol. 2012, 9, 510–519. [Google Scholar] [CrossRef]
  98. Honda, T.; Ando, M.; Ando, J.; Ishii, M.; Sakiyama, Y.; Ohara, K.; Toyota, T.; Ohtaka, M.; Masuda, A.; Terao, Y.; et al. Sustainable Tumor-Suppressive Effect of iPSC-Derived Rejuvenated T Cells Targeting Cervical Cancers. Mol. Ther. 2020, 28, 2394–2405. [Google Scholar] [CrossRef]
  99. Kashima, S.; Maeda, T.; Masuda, K.; Nagano, S.; Inoue, T.; Takeda, M.; Kono, Y.; Kobayashi, T.; Saito, S.; Higuchi, T.; et al. Cytotoxic T Lymphocytes Regenerated from iPS Cells Have Therapeutic Efficacy in a Patient-Derived Xenograft Solid Tumor Model. iScience 2020, 23, 100998. [Google Scholar] [CrossRef]
  100. Ishii, M.; Ando, J.; Yamazaki, S.; Toyota, T.; Ohara, K.; Furukawa, Y.; Suehara, Y.; Nakanishi, M.; Nakashima, K.; Ohshima, K.; et al. iPSC-Derived Neoantigen-Specific CTL Therapy for Ewing Sarcoma. Cancer Immunol. Res. 2021, 9, 1175–1186. [Google Scholar] [CrossRef]
  101. Ando, M.; Nakauchi, H. ‘Off-the-shelf’ immunotherapy with iPSC-derived rejuvenated cytotoxic T lymphocytes. Exp. Hematol. 2017, 47, 2–12. [Google Scholar] [CrossRef]
  102. Lee, S.J.; Klein, J.; Haagenson, M.; Baxter-Lowe, L.A.; Confer, D.L.; Eapen, M.; Fernandez-Vina, M.; Flomenberg, N.; Horowitz, M.; Hurley, C.K.; et al. High-resolution donor-recipient HLA matching contributes to the success of unrelated donor marrow transplantation. Blood 2007, 110, 4576–4583. [Google Scholar] [CrossRef] [Green Version]
  103. June, C.H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379, 64–73. [Google Scholar] [CrossRef]
  104. Lim, W.A.; June, C.H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168, 724–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Rafiq, S.; Hackett, C.S.; Brentjens, R.J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 2020, 17, 147–167. [Google Scholar] [CrossRef]
  106. Jackson, H.J.; Rafiq, S.; Brentjens, R.J. Driving CAR T-cells forward. Nat. Rev. Clin. Oncol. 2016, 13, 370–383. [Google Scholar] [CrossRef]
  107. Brudno, J.N.; Kochenderfer, J.N. Chimeric antigen receptor T-cell therapies for lymphoma. Nat. Rev. Clin. Oncol. 2018, 15, 31–46. [Google Scholar] [CrossRef]
  108. Kershaw, M.H.; Westwood, J.A.; Darcy, P.K. Gene-engineered T cells for cancer therapy. Nat. Rev. Cancer 2013, 13, 525–541. [Google Scholar] [CrossRef] [PubMed]
  109. Kershaw, M.H.; Westwood, J.A.; Parker, L.L.; Wang, G.; Eshhar, Z.; Mavroukakis, S.A.; White, D.E.; Wunderlich, J.R.; Canevari, S.; Rogers-Freezer, L.; et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer Res. 2006, 12, 6106–6115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Wang, L.X.; Westwood, J.A.; Moeller, M.; Duong, C.P.; Wei, W.Z.; Malaterre, J.; Trapani, J.A.; Neeson, P.; Smyth, M.J.; Kershaw, M.H.; et al. Tumor ablation by gene-modified T cells in the absence of autoimmunity. Cancer Res. 2010, 70, 9591–9598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Savoldo, B.; Ramos, C.A.; Liu, E.; Mims, M.P.; Keating, M.J.; Carrum, G.; Kamble, R.T.; Bollard, C.M.; Gee, A.P.; Mei, Z.; 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. [Google Scholar] [CrossRef] [Green Version]
  112. Kochenderfer, J.N.; Rosenberg, S.A. Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat. Rev. Clin. Oncol. 2013, 10, 267–276. [Google Scholar] [CrossRef] [PubMed]
  113. Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R.; Teachey, D.T.; Chew, A.; Hauck, B.; Wright, J.F.; et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [Green Version]
  114. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; 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. [Google Scholar] [CrossRef]
  115. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
  116. Turtle, C.J.; Hanafi, L.A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Investig. 2016, 126, 2123–2138. [Google Scholar] [CrossRef] [Green Version]
  117. Shah, N.N.; Fry, T.J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 2019, 16, 372–385. [Google Scholar] [CrossRef] [PubMed]
  118. Orlando, E.J.; Han, X.; Tribouley, C.; Wood, P.A.; Leary, R.J.; Riester, M.; Levine, J.E.; Qayed, M.; Grupp, S.A.; Boyer, M.; et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat. Med. 2018, 24, 1504–1506. [Google Scholar] [CrossRef]
  119. Majzner, R.G.; Mackall, C.L. Tumor Antigen Escape from CAR T-cell Therapy. Cancer Discov. 2018, 8, 1219–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef] [PubMed]
  121. Schuster, S.J.; Svoboda, J.; Chong, E.A.; Nasta, S.D.; Mato, A.R.; Anak, Ö.; Brogdon, J.L.; Pruteanu-Malinici, I.; Bhoj, V.; Landsburg, D.; et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N. Engl. J. Med. 2017, 377, 2545–2554. [Google Scholar] [CrossRef]
  122. Themeli, M.; Kloss, C.C.; Ciriello, G.; Fedorov, V.D.; Perna, F.; Gonen, M.; Sadelain, M. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 2013, 31, 928–933. [Google Scholar] [CrossRef]
  123. Themeli, M.; Rivière, I.; Sadelain, M. New cell sources for T cell engineering and adoptive immunotherapy. Cell Stem Cell 2015, 16, 357–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Harada, S.; Ando, M.; Ando, J.; Ishii, M.; Yamaguchi, T.; Yamazaki, S.; Toyota, T.; Ohara, K.; Ohtaka, M.; Nakanishi, M.; et al. Dual-antigen targeted iPSC-derived chimeric antigen receptor-T cell therapy for refractory lymphoma. Mol. Ther. 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  125. Hinrichs, C.S.; Restifo, N.P. Reassessing target antigens for adoptive T-cell therapy. Nat. Biotechnol. 2013, 31, 999–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. 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. [Google Scholar] [CrossRef] [PubMed]
  127. Bonini, C.; Ferrari, G.; Verzeletti, S.; Servida, P.; Zappone, E.; Ruggieri, L.; Ponzoni, M.; Rossini, S.; Mavilio, F.; Traversari, C.; et al. HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia. Science 1997, 276, 1719–1724. [Google Scholar] [CrossRef] [PubMed]
  128. Ciceri, F.; Bonini, C.; Gallo-Stampino, C.; Bordignon, C. Modulation of GvHD by suicide-gene transduced donor T lymphocytes: Clinical applications in mismatched transplantation. Cytotherapy 2005, 7, 144–149. [Google Scholar] [CrossRef]
  129. Berger, C.; Flowers, M.E.; Warren, E.H.; Riddell, S.R. Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation. Blood 2006, 107, 2294–2302. [Google Scholar] [CrossRef] [Green Version]
  130. Brenner, M.K.; Gottschalk, S.; Leen, A.M.; Vera, J.F. Is cancer gene therapy an empty suit? Lancet Oncol. 2013, 14, e447–e456. [Google Scholar] [CrossRef] [Green Version]
  131. Hoyos, V.; Savoldo, B.; Quintarelli, C.; Mahendravada, A.; Zhang, M.; Vera, J.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; Dotti, G. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia 2010, 24, 1160–1170. [Google Scholar] [CrossRef] [Green Version]
  132. Straathof, K.C.; Pulè, M.A.; Yotnda, P.; Dotti, G.; Vanin, E.F.; Brenner, M.K.; Heslop, H.E.; Spencer, D.M.; Rooney, C.M. An inducible caspase 9 safety switch for T-cell therapy. Blood 2005, 105, 4247–4254. [Google Scholar] [CrossRef]
  133. Clackson, T.; Yang, W.; Rozamus, L.W.; Hatada, M.; Amara, J.F.; Rollins, C.T.; Stevenson, L.F.; Magari, S.R.; Wood, S.A.; Courage, N.L.; et al. Redesigning an FKBP-ligand interface to generate chemical dimerizers with novel specificity. Proc. Natl. Acad. Sci. USA 1998, 95, 10437–10442. [Google Scholar] [CrossRef] [Green Version]
  134. Di Stasi, A.; Tey, S.K.; Dotti, G.; Fujita, Y.; Kennedy-Nasser, A.; Martinez, C.; Straathof, K.; Liu, E.; Durett, A.G.; Grilley, B.; et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 2011, 365, 1673–1683. [Google Scholar] [CrossRef] [Green Version]
  135. Ando, M.; Hoyos, V.; Yagyu, S.; Tao, W.; Ramos, C.A.; Dotti, G.; Brenner, M.K.; Bouchier-Hayes, L. Bortezomib sensitizes non-small cell lung cancer to mesenchymal stromal cell-delivered inducible caspase-9-mediated cytotoxicity. Cancer Gene Ther. 2014, 21, 472–482. [Google Scholar] [CrossRef] [Green Version]
  136. Zhou, X.; Di Stasi, A.; Tey, S.K.; Krance, R.A.; Martinez, C.; Leung, K.S.; Durett, A.G.; Wu, M.F.; Liu, H.; Leen, A.M.; et al. Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood 2014, 123, 3895–3905. [Google Scholar] [CrossRef] [Green Version]
  137. Zhou, X.; Dotti, G.; Krance, R.A.; Martinez, C.A.; Naik, S.; Kamble, R.T.; Durett, A.G.; Dakhova, O.; Savoldo, B.; Di Stasi, A.; et al. Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation. Blood 2015, 125, 4103–4113. [Google Scholar] [CrossRef] [Green Version]
  138. Zhou, X.; Brenner, M.K. Improving the safety of T-Cell therapies using an inducible caspase-9 gene. Exp. Hematol. 2016, 44, 1013–1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Wu, C.; Hong, S.G.; Winkler, T.; Spencer, D.M.; Jares, A.; Ichwan, B.; Nicolae, A.; Guo, V.; Larochelle, A.; Dunbar, C.E. Development of an inducible caspase-9 safety switch for pluripotent stem cell-based therapies. Mol. Ther. Methods Clin. Dev. 2014, 1, 14053. [Google Scholar] [CrossRef] [PubMed]
  140. Barese, C.N.; Felizardo, T.C.; Sellers, S.E.; Keyvanfar, K.; Di Stasi, A.; Metzger, M.E.; Krouse, A.E.; Donahue, R.E.; Spencer, D.M.; Dunbar, C.E. Regulated apoptosis of genetically modified hematopoietic stem and progenitor cells via an inducible caspase-9 suicide gene in rhesus macaques. Stem Cells 2015, 33, 91–100. [Google Scholar] [CrossRef] [Green Version]
  141. Yagyu, S.; Hoyos, V.; Del Bufalo, F.; Brenner, M.K. An Inducible Caspase-9 Suicide Gene to Improve the Safety of Therapy Using Human Induced Pluripotent Stem Cells. Mol. Ther. 2015, 23, 1475–1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Itakura, G.; Kawabata, S.; Ando, M.; Nishiyama, Y.; Sugai, K.; Ozaki, M.; Iida, T.; Ookubo, T.; Kojima, K.; Kashiwagi, R.; et al. Fail-Safe System against Potential Tumorigenicity after Transplantation of iPSC Derivatives. Stem Cell Rep. 2017, 8, 673–684. [Google Scholar] [CrossRef]
  143. Martin, R.M.; Fowler, J.L.; Cromer, M.K.; Lesch, B.J.; Ponce, E.; Uchida, N.; Nishimura, T.; Porteus, M.H.; Loh, K.M. Improving the safety of human pluripotent stem cell therapies using genome-edited orthogonal safeguards. Nat. Commun. 2020, 11, 2713. [Google Scholar] [CrossRef]
  144. Heczey, A.; Louis, C.U.; Savoldo, B.; Dakhova, O.; Durett, A.; Grilley, B.; Liu, H.; Wu, M.F.; Mei, Z.; Gee, A.; et al. CAR T Cells Administered in Combination with Lymphodepletion and PD-1 Inhibition to Patients with Neuroblastoma. Mol. Ther. 2017, 25, 2214–2224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Cruz, C.R.; Micklethwaite, K.P.; Savoldo, B.; Ramos, C.A.; Lam, S.; Ku, S.; Diouf, O.; Liu, E.; Barrett, A.J.; Ito, S.; 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. [Google Scholar] [CrossRef]
  146. Deuse, T.; Hu, X.; Gravina, A.; Wang, D.; Tediashvili, G.; De, C.; Thayer, W.O.; Wahl, A.; Garcia, J.V.; Reichenspurner, H.; et al. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat. Biotechnol. 2019, 37, 252–258. [Google Scholar] [CrossRef]
  147. Ikeda, K.; Uchida, N.; Nishimura, T.; White, J.; Martin, R.M.; Nakauchi, H.; Sebastiano, V.; Weinberg, K.I.; Porteus, M.H. Efficient scarless genome editing in human pluripotent stem cells. Nat. Methods 2018, 15, 1045–1047. [Google Scholar] [CrossRef] [PubMed]
  148. Xu, H.; Wang, B.; Ono, M.; Kagita, A.; Fujii, K.; Sasakawa, N.; Ueda, T.; Gee, P.; Nishikawa, M.; Nomura, M.; et al. Targeted Disruption of HLA Genes via CRISPR-Cas9 Generates iPSCs with Enhanced Immune Compatibility. Cell Stem Cell 2019, 24, 566–578.e567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Suzue, K.; Reinherz, E.L.; Koyasu, S. Critical role of NK but not NKT cells in acute rejection of parental bone marrow cells in F1 hybrid mice. Eur. J. Immunol. 2001, 31, 3147–3152. [Google Scholar] [CrossRef]
  150. Masuda, K.; Kawamoto, H. Possible NK cell-mediated immune responses against iPSC-derived cells in allogeneic transplantation settings. Inflamm. Regen. 2021, 41, 2. [Google Scholar] [CrossRef] [PubMed]
  151. Gornalusse, G.G.; Hirata, R.K.; Funk, S.E.; Riolobos, L.; Lopes, V.S.; Manske, G.; Prunkard, D.; Colunga, A.G.; Hanafi, L.A.; Clegg, D.O.; et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat. Biotechnol. 2017, 35, 765–772. [Google Scholar] [CrossRef] [Green Version]
  152. Han, X.; Wang, M.; Duan, S.; Franco, P.J.; Kenty, J.H.; Hedrick, P.; Xia, Y.; Allen, A.; Ferreira, L.M.R.; Strominger, J.L.; et al. Generation of hypoimmunogenic human pluripotent stem cells. Proc. Natl. Acad. Sci. USA 2019, 116, 10441–10446. [Google Scholar] [CrossRef] [Green Version]
  153. Fu, B.; Zhou, Y.; Ni, X.; Tong, X.; Xu, X.; Dong, Z.; Sun, R.; Tian, Z.; Wei, H. Natural Killer Cells Promote Fetal Development through the Secretion of Growth-Promoting Factors. Immunity 2017, 47, 1100–1113.e1106. [Google Scholar] [CrossRef] [Green Version]
  154. Zhao, H.X.; Jiang, F.; Zhu, Y.J.; Wang, L.; Li, K.; Li, Y.; Wang, X.H.; Li, L.S.; Yao, Y.Q. Enhanced Immunological Tolerance by HLA-G1 from Neural Progenitor Cells (NPCs) Derived from Human Embryonic Stem Cells (hESCs). Cell Physiol. Biochem. 2017, 44, 1435–1444. [Google Scholar] [CrossRef]
  155. Zhao, L.; Teklemariam, T.; Hantash, B.M. Heterelogous expression of mutated HLA-G decreases immunogenicity of human embryonic stem cells and their epidermal derivatives. Stem Cell Res 2014, 13, 342–354. [Google Scholar] [CrossRef] [Green Version]
  156. Cudkowicz, G.; Stimpfling, J.H. Deficient growth of c57bl marrow cells transplanted in f1 hybrid mice. association with the histocompatibility-2 locus. Immunology 1964, 7, 291–306. [Google Scholar] [PubMed]
  157. Raulet, D.H. Bone Marrow Cell Rejection, MHC, NK Cells, and Missing Self Recognition: Ain’t That Peculiar (with Apologies to Marvin Gaye). J. Immunol. 2015, 195, 2923–2925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Murphy, W.J.; Kumar, V.; Bennett, M. Rejection of bone marrow allografts by mice with severe combined immune deficiency (SCID). Evidence that natural killer cells can mediate the specificity of marrow graft rejection. J. Exp. Med. 1987, 165, 1212–1217. [Google Scholar] [CrossRef] [PubMed]
  159. Wang, B.; Iriguchi, S.; Waseda, M.; Ueda, N.; Ueda, T.; Xu, H.; Minagawa, A.; Ishikawa, A.; Yano, H.; Ishi, T.; et al. Generation of hypoimmunogenic T cells from genetically engineered allogeneic human induced pluripotent stem cells. Nat. Biomed. Eng. 2021, 5, 429–440. [Google Scholar] [CrossRef]
Figure 1. Autologous CART therapy and iPSC-derived engineered T cell therapy. (A) T cells are extracted from patients by apheresis and CARs are integrated into T cells, then re-infused to the patient. (B) After T cells are reprogrammed into T-iPSCs, T-iPSCs can be transduced with CARs, TCRs, suicide gene iC9, etc.
Figure 1. Autologous CART therapy and iPSC-derived engineered T cell therapy. (A) T cells are extracted from patients by apheresis and CARs are integrated into T cells, then re-infused to the patient. (B) After T cells are reprogrammed into T-iPSCs, T-iPSCs can be transduced with CARs, TCRs, suicide gene iC9, etc.
Cells 11 00269 g001
Figure 2. Expression of suicide genes in T lymphocytes. (A) HSV-TK phosphorylates ganciclovir (GCV) to nucleoside monophosphate, then a second cellular kinase phosphorylates this into nucleoside triphosphate. This inhibits DNA synthesis, resulting in death of dividing cells. (B) In the mitochondrial apoptosis pathway, translocation of activated BAX or BID to the mitochondria creates pores and allows cytochrome C to move to the cytosol. Cytochrome C then activates caspase-9, resulting in the activation of caspase-3, which leads to apoptosis. iC9 was made by the fusion of human caspase-9 to a modified FK506-binding protein (FKBP12), allowing dimerization. By adding CID, iC9 is dimerized and activates caspase-3 directly, also inducing apoptosis.
Figure 2. Expression of suicide genes in T lymphocytes. (A) HSV-TK phosphorylates ganciclovir (GCV) to nucleoside monophosphate, then a second cellular kinase phosphorylates this into nucleoside triphosphate. This inhibits DNA synthesis, resulting in death of dividing cells. (B) In the mitochondrial apoptosis pathway, translocation of activated BAX or BID to the mitochondria creates pores and allows cytochrome C to move to the cytosol. Cytochrome C then activates caspase-9, resulting in the activation of caspase-3, which leads to apoptosis. iC9 was made by the fusion of human caspase-9 to a modified FK506-binding protein (FKBP12), allowing dimerization. By adding CID, iC9 is dimerized and activates caspase-3 directly, also inducing apoptosis.
Cells 11 00269 g002
Figure 3. CRISPR/Cas9 gene editing of iPSCs can reduce immune rejection by recipient cells. Deletion of HLA class I by B2M knock out prevents T cell rejection. HLA-C and HLA-E protect against NK cell “missing self” response. NK cell attack can also be blocked by the overexpression of CD47.
Figure 3. CRISPR/Cas9 gene editing of iPSCs can reduce immune rejection by recipient cells. Deletion of HLA class I by B2M knock out prevents T cell rejection. HLA-C and HLA-E protect against NK cell “missing self” response. NK cell attack can also be blocked by the overexpression of CD47.
Cells 11 00269 g003
Table 1. Various strategies in allogeneic CART therapy.
Table 1. Various strategies in allogeneic CART therapy.
CompanyProductsCARAllogeneic Cell SourceStrategy to Evade GVHD or Immune RejectionTarget GeneClinical Trial Identifier
Allogene TherapeuticsALLO-501 (in USA)CD19T cellTALENTRAC and CD52NCT03939026
ALLO-715BCMANCT04093596
ALLO-316CD70NCT04696731
Adicet Bio, Inc.ADI-001CD20γδT cell NCT04735471
Caribou Biosciences, Inc.CB010ACD19T cellCRISPR/Cas9TRAC, PD1 (KO)NCT04637763
CellectisUCART-123CD123T cellTALENTRAC (KO)NCT03190278 (AML)
NCT03203369 (BPDCN)
NCT04150497
NCT04142619
UCART-22CD22TRAC and CD52 (KO)
UCART-CS1CS1TRAC and CS1 (KO)
CelyadCYAD-211BCMAT cellshRNACD3ζNCT03692429
CYAD-101NKG2DLOverexpression of TIM-NCT03692429
CRISPR Therapeutics AGCTX110CD19T cellCRISPR/Cas9TRAC and B2M (KO)NCT04035434
CTX120BCMANCT04244656
CTX130CD70NCT04502446
Fate TherapeuticsFT-819CD19iPSC-derived T cellCRISPR/Cas9TRACNCT04629729
Great Ormond Street Hospital for ChildrenPBLTT52CAR19CD19T cellCRISPR/Cas9CD52 and TRAC (KO)NCT04557436
Kurr therapeuticsKUR-502CD19NKT NCT03774654
Precision BiosciencesPBCAR0191CD19T cellmeganuclease mRNA
shRNA
B2M (shRNA), HLA-ENCT03666000
PBCAR19BCD19NCT04649112
PBCAR20ACD20NCT04030195
PBCAR269ABCMANCT04171843
Tessa Therapeutics Pte Ltd.CD30.CAR-EBVST cellsCD30EBVST NCT04288726
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Furukawa, Y.; Hamano, Y.; Shirane, S.; Kinoshita, S.; Azusawa, Y.; Ando, J.; Nakauchi, H.; Ando, M. Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using iPSC Technology and Gene Editing. Cells 2022, 11, 269. https://doi.org/10.3390/cells11020269

AMA Style

Furukawa Y, Hamano Y, Shirane S, Kinoshita S, Azusawa Y, Ando J, Nakauchi H, Ando M. Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using iPSC Technology and Gene Editing. Cells. 2022; 11(2):269. https://doi.org/10.3390/cells11020269

Chicago/Turabian Style

Furukawa, Yoshiki, Yasuharu Hamano, Shuichi Shirane, Shintaro Kinoshita, Yoko Azusawa, Jun Ando, Hiromitsu Nakauchi, and Miki Ando. 2022. "Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using iPSC Technology and Gene Editing" Cells 11, no. 2: 269. https://doi.org/10.3390/cells11020269

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop