Abstract
Allogeneic hematopoietic stem cell transplantation is an effective therapy for high-risk leukemias. In children, graft manipulation based on the selective removal of αβ T cells and B cells has been shown to reduce the risk of acute and chronic graft-versus-host disease, thus allowing the use of haploidentical donors which expands the population that allogeneic hematopoietic stem cell transplantation can be used in. Leukemic relapse, however, remains a problem. T cells expressing chimeric antigen receptors can potently eliminate leukemia, including in the central nervous system. We hypothesized that by modifying donor αβ T cells to simultaneously express a CD19-specific chimeric antigen receptors and inactivating the T cell receptor by genome editing, we could create a therapy that enhances the anti-leukemic efficacy of the stem cell transplant without increasing the risk of graft-versus-host disease. Using genome editing with Cas9 ribonucleoprotein and adeno-associated virus serotype 6, we integrate a CD19-specific chimeric antigen receptor in-frame into the TRAC locus. Greater than 90% of cells lost TCR expression, while >75% expressed the CAR. The product was further purified to ultimately have less than 0.05% residual TCR+ cells. In vitro, the CAR T cells efficiently eliminated target cells and produced high cytokine levels when challenged with CD19+ leukemia cells. In vivo, the gene modified T cells eliminated leukemia without causing xenogeneic graft-versus-host disease in a xenograft model. Gene editing was highly specific with no evidence of off-target effects. These data support the concept that the addition of αβ T cell-derived, genome edited T cells expressing CD19-specific chimeric antigen receptors could enhance the anti-leukemic efficacy of αβ T cell-depleted haploidentical hematopoietic stem cell transplantation without increasing the risk of graft-versus-host disease.Introduction
Despite improvements over the last decades, treatment outcomes remain unfavorable in pediatric patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia.1,2 Allogeneic hematopoietic stem cell transplantation (HSCT) has been successfully employed to treat high-risk leukemias,3 providing a graft-versus- leukemia4 effect but also carrying the risk of graft-versus-host disease (GvHD). In order to overcome the challenge of limited donor availability, 6 innovative protocols have been developed that allow the use of grafts from haploidentical donors. A successful strategy for haploidentical transplantation is based on the selective elimination of αβT cells and CD19+ B cells (αβhaplo-HSCT),7 which is associated with a very low incidence of transplantation-related mortality and GvHD (Figure 1A).8-10 In contrast to CD34+-cell selection, this method of manipulation allows the transfer not only of donor hematopoietic stem cells to the recipient, but also committed hematopoietic progenitors as well as mature natural killer and γδT cells,11,12 which may provide a protective effect against leukemia relapse and reduce the risk of infectious complications.13 With significant improvement in non-relapse mortality, disease relapse has become the most important cause of treatment failure in patients with malignancies undergoing αβhaplo- HSCT.8 In particular, the outcome of αβhaplo-HSCT in children with leukemia not in complete remission or beyond second complete remission has been poor.9,14 For this reason, it is necessary to develop novel strategies to reduce leukemic relapse after haplo-HSCT, without increasing the incidence of GvHD or transplant-related mortality.
An intriguing approach to reducing leukemic relapse is to follow haplo-HSCT with subsequent anti-leukemic cell therapy15 derived from the stem cell donor (Figure 1AD), since these cells are from healthy immune systems and are also syngeneic (functionally autologous) to the donor graft, rendering them resistant to immune rejection after transplantation. While the infusion of donor-derived T cells (donor lymphocyte infusion) has been used in various contexts to enhance antileukemic efficacy (Figure 1B), it is accompanied by a high risk of severe GvHD.16-22 An improvement over donor lymphocyte infusion is to genetically engineer the donor T cells with a safety switch (suicide gene) such that the cells can be quickly eliminated if severe GvHD occurs (Figure 1C). Early trials have suggested that this strategy does help to prevent relapse and the suicide switch (inducible caspase 9) is effective at eliminating alloreactive cells if GvHD occurs.23-25 Although this strategy allows GvHD to be controlled after it occurs, the benefit of the graft-versusleukemia effect and the risk of GvHD remain linked to each other. It would be an improvement, therefore, to establish an approach that provides anti-leukemic activity without GvHD.
Chimeric antigen receptors (CAR) can redirect T-cell cytotoxicity towards cancer-related antigens and achieve remissions in otherwise refractory hematologic malignancies expressing these targets.26,27 Currently, the most commonly used CAR T-cell products are manufactured from patient-derived autologous T cells that are harvested and transduced with a semi-randomly integrating viral vector for delivery and expression of the CAR gene, and then infused back into the patient after lymphodepleting therapy. 28 This is associated with high variability in the CAR T-cell product and manufacturing failures. Furthermore, contaminations of the autologous cells with leukemic cells29 and the risk of insertional mutagenesis associated with randomly integrating viral vectors30 are challenges associated with the established approach.
We hypothesized that αβhaplo-HSCT in combination with CAR T cells generated through genome editing of donor-derived T cells could provide the foundation for an optimal approach that addresses these challenges (Figure 1D). We here show that the T-cell receptor (TCR) αβ+/CD19+ cell fraction that is removed from the graft can be used to engineer non-alloreactive CAR T cells through homologous recombination-mediated genome editing by targeted integration of a CD19-specific CAR in-frame into the TCR alpha chain (TRAC) locus (“αβTCR-CD19 CAR-T”), and demonstrate the antileukemic efficacy of this product in vitro and in vivo. This novel and innovative approach allows for the creation of two different cellular immunotherapy products, with complementary antileukemic mechanisms, from a single apheresis: the αβ haplo-HSCT which provides donor-derived natural killer cells, γδ T cells and the human leukocyte antigen (HLA)- dependent activity of polyclonal T cells, while the “leftover” cell fraction is salvaged to become a therapeutic CAR T-cell product with an HLA-independent mechanism and potential to improve cure rates without causing GvHD.
Methods
Plasmid cloning and adeno-associated virus production
Transfer plasmids were cloned between the inverted terminal repeat sequences in pAAV-MCS (Agilent Technologies). The CAR comprises a GM-CSFRα leader sequence, the FMC63 scFv,31 CD28 hinge, transmembrane and intracellular sequences and the CD3ξ intracellular domain. Recombinant adeno-associated virus serotype 6 (rAAV6) production and titration are described in the Online Supplementary Methods.
Apheresis and cell processing
αβhaplo-HSCT donors received granulocyte-colony stimulating factor for 4 days at the total dose of 16 μg/kg body weight and apheresis was performed on the fifth day. If the CD34+ cell count was <40/μL on day 4, a CXCR4 antagonist (plerixafor, Mozobil) was given. Manipulations were performed in a closed system according to Good Manufacturing Practice (GMP) standards with clinical grade reagents and instruments from Miltenyi Biotec (Bergisch Gladbach, Germany).
T-cell culture and genome editing
All human cells were handled according to a protocol approved by the Institutional Review Board at Stanford University. The TCRαβ+/CD19+ cell fraction (non-target fraction from the graft manipulation procedure) was used Fresh or cryo - preserved. T cells were activated for 3 days and beads removed before electroporation. Electroporation and gene targeting were performed as previously described.32
Depletion by magnetic bead activated cell sorting
Depletion of TCRαβ+ cells was performed using reagents from Miltenyi according to the manufacturer’s instructions, except after coating with the Streptavidin-microbeads, when the cells were diluted and passed through the column without the washing step. For details see the Online Supplementary Methods.
In vitro cytokine measurement and cytotoxicity assay
CD19+ Nalm6-GL cells or CD19+ Raji cells (GFP-Luc+) were used in co-culture assays with the CAR T cells or control T cells to determine interleukin-2 and interferon-production of the CAR T cells and cytotoxicity. For details see the Online Supplementary Methods.
In vivo xenograft assay
All experiments involving mice were performed according to a protocol approved by the Administrative Panel on Laboratory Animal Care at Stanford University. CD19+ Nalm6-GL cells (5x105) were transplanted intravenously (i.v.) into 6- to 12-week old male NSG mice. Four days later, tumor burden was evaluated by IVIS bioluminescence imaging (PerkinElmer) and the indicated numbers of CAR T cells or control cells injected i.v.. Tumor burden was followed weekly by IVIS imaging.
Antibodies used for flow cytometry
NGFR-APC, NGFR-PE, TCRαβ-FITC, CD19-A488, CD19- A700, CD62L-BV421, CD45RA-PE, CD4-PerCP-Cy5.5, CD8a- APC-Cy7 (all from Biolegend) were used. For CD45RA/CD62L staining, isotype controls as recommended by the manufacturer were used to determine positive and negative populations. The APC-conjugated CD19-CAR idiotype antibody was a gift from Crystal Mackall.
Off-target analysis
sgRNA target sites were identified and their specificity score calculated by bioinformatics (crispor.tefor.net33). COSMID (crispr.bme.gatech.edu34) was used to identify potential off-target sites in the human genome. For analysis of predicted off-targets, gene editing or mock treatment was performed on T cells from six different donors and predicted off-target sites sequenced using an Illumina MiSeq as described previously.35 For details see the Online Supplementary Methods.
Statistics
Plots show means with error bars representing either the standard deviation or 95% confidence interval (95% CI), as indicated. Groups were compared by statistical tests as described in the figure legends using Prism 7 (GraphPad). Asterisks indicate statistical significance: *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001. All t tests are two-tailed.
Results
Genome editing on TCRαβ+ cells depleted from the graft during αβhaploidentical hematopoietic stem cell transplanation to create chimeric antigen receptor T cells
We prospectively collected the TCRαβ+/CD19+ cell fraction (non-target fraction) removed from grafts during αβhaplo-HSCT procedures. It has recently been shown that homologous recombination-mediated genome editing using Cas9 ribonucleoprotein (RNP) and AAV6 can mediate targeted integration of a CAR into the TRAC locus,36,37 with up to 50% of cells expressing the CAR. This approach offers the advantages that it establishes TCR knockout in the majority of CAR+ cells, avoids the risk of insertional mutagenesis of randomly-integrating viral vectors, and allows the cells to modulate CAR expression if the CAR is integrated in-frame into the endogenous locus.37
We used homologous recombination-based genome editing32 to integrate a CD19.28.ξ-CAR in-frame into the open-reading frame of the TRAC locus (Figures 1D and 2A), similarly to a recently described approach.37 Disruption of TRAC leads to loss of expression of the TCR complex on the cell surface.38 TRAC is advantageous over TRBC because it only exists once per haploid genome. Although 4-1BB co-stimulation has been shown to lead to enhanced persistence of CAR T cells due to decreased exhaustion,39 we chose CD28 as the co-stimulatory domain in the CAR in order to account for the expected low number of target cells in the setting after HSCT. In the presence of minimal disease burden and absent or low numbers of B cells immediately after HSCT, the stronger effector signaling from CD28 costimulation could lead to enhanced activation and proliferation of the CAR T cells. It was recently shown that the method of targeted integration of a CAR into the TRAC locus with expression from the endogenous promotor can preserve functionality of cells with CD28 costimulation, 37 which are otherwise prone to exhaustion.
We used an sgRNA (termed TRAC-1) that had previously been shown to have high on-target activity and no detectable off-target activity.40 Potential off-target sites across the human genome were predicted by the COSMID algorithm34 and comparison to other possible sgRNA (Online Supplementary Figure S1A, B) confirmed that this sgRNA was among the most specific in exon 1 of TRAC with no highly similar off-targets. The most similar predicted off-target site had three mismatched nucleotides, suggesting a low probability of Cas9 cleavage activity (see below).
To investigate whether genome editing with TCR knockout and targeted integration of a CAR is feasible in TCRαβ+ T cells removed from the graft during αβhaplo- HSCT, we cultured and stimulated these cells and electroporated them with an RNP complex consisting of a highfidelity version of the Cas9 protein41 complexed with chemically-modified sgRNA,42 immediately followed by transduction of a DNA repair template by a non-integrating rAAV6. Following this process, on average 95.7% of the cells lost TCR expression (95% CI: 94.2-97.3) and 79.4% (95% CI: 73.5-85.3) of bulk cells and 81.4% (95% CI: 75.7-87.1) of TCR- cells expressed tNGFR (a truncated non-signaling cell surface form of the nerve growth factor receptor which has been used safely in clinical immunotherapy trials24) (Figure 2B and C). These unprecedented efficiencies of targeted integration of a large gene expression cassette (2.7 kb) in primary T cells was reproduced in cells from 11 different donors with similar efficiencies (Figure 2C). Importantly, this proves that cellular double-strand break repair can efficiently be skewed toward homologous recombination to the level at which it constitutes the predominant repair pathway and targeted integration becomes more frequent than insertion/deletion formation by non-homologous endjoining. Notably, the starting cells had been processed at two different GMP facilities (5 at the University of California, San Francisco and 6 at the Stanford University Laboratory for Cell and Gene Medicine), but the outcome after gene editing was highly reproducible (Figure 2C). To confirm co-expression of the CAR in the NGFR+ cells, we stained the cells with an antibody that detects the CAR, which confirmed that both genes of the bicistronic expression cassette are translated (Figure 2D).
Efficient depletion of potentially alloreactive TCR+ cells and optimization of editing methods
Despite the efficiency of the genome editing process, a small fraction of cells (<8%) retained expression of their TCR. Prior studies have suggested that the frequency of GvHD occurrence for allogeneic CAR T cells with CD28 co-stimulation is low,43 supposedly due to exhaustion and clonal deletion of alloreactive cells44 stimulated through both the CAR and their TCR. Despite these promising results, this is not guaranteed to be universally true, especially since our method creates CAR expression levels different from virally transduced CAR T cells and could lead to different biological properties. The residual TCR+ cells – being HLA-haploidentical to the recipient - carry the potential for alloreactivity, and their further depletion from the cell product could decrease the probability of GvHD and allow higher doses of cells to be administered. We therefore evaluated the depletion of residual TCRαβ+ cells from the expanded cell population by magnetic bead activated cell sorting using reagents for which GMP-compatible counterparts are available. We were able to achieve efficient depletion with a maximum of 0.03% TCRαβ+ cells remaining in the resulting cell product (a depletion efficiency of 2-3 orders of magnitude) (Figure 2E and F), a higher efficiency than in prior studies that created TCR– CAR T cells.45,46We termed the resulting cell product after genome editing, expansion and TCRαβ+ depletion “αβTCR-CD19 CAR-T”.
The cells rapidly expanded following genome editing (over 60-fold in 7 days), with no negative effect of RNP electroporation on cell yields, an 11% decrease in expansion after AAV transduction, and a decrease of 27% for cells electroporated with RNP and transduced with AAV (Online Supplementary Figure S2A). This suggested that AAV transduction is the main factor affecting cell expansion, which led us to determine the optimal AAV dose for maximal gene targeting efficiency. Interestingly, we found that a change in the multiplicity of infection beyond 2,500 vector genomes (vg)/cell only led to a minor change in targeting outcomes with saturation at 5,000 vg/cell (Online Supplementary Figure S2B). Instead, the duration of time during which the cells were kept at a high concentration for AAV transduction (>5x106 cells/mL) directly after electroporation and before dilution to the target cell density influenced gene targeting outcomes to a greater extent (Online Supplementary Figure S2C). Using a multiplicity of infection of 5,000 vg/cell and a prolonged transduction time at high density (>12 h), we observed that the cells expanded on average 103-fold within the 7 days following gene editing (Figure 2G). With these conditions for gene editing, the expansion rate of the cells was primarily dependent on the culture density, reaching the threshold of >100-fold expansion in 7 days if cultured at 0.125x106 cells/mL or within 10 days if cultured at 0.5x106 cells/mL (Figure 2H). This confirms that the CAR T cells are able to expand rapidly without further TCR stimulation after gene editing despite the manipulation during the gene editing process. This will aid in the development of a cell product at clinically relevant scale.
To summarize, we achieved efficient disruption of the TCR and high frequencies of CAR expression in T cells derived from the otherwise discarded TCRαβ+ T cells, while allowing for rapid expansion of the resulting cells after the editing process when using an optimized protocol.
Phenotype and in vitro efficacy
To measure in vitro cytokine production and cytotoxic activity, we used the CD19+ lymphoblastic cell lines Nalm6-GL and Raji (GFP-Luciferase transduced) as target cells. After co-culture for 20 h, we were able to measure production of interleukin-2 and interferon-γ in the cell culture supernatant specifically for cells with integration of the CAR (Figure 3A). To estimate the fraction of target cells that was killed, we determined the counts of target cells (identified by their GFP expression) after 20 h of coculture with either CAR T cells or control cells relative to samples cultured without effector cells, which showed cytotoxic activity even at low effector-to-target (E:T) ratios (Figure 3B).
To determine the fate of the B cells, which are part of the source cell population besides the αβT cells, we followed the CD19+ cell population by phenotyping and were able to detect their disappearance as early as 24 h after gene targeting (2.53%, vs. 7.9-11.8% in the control condition in which the T cells did not express CAR), suggesting early cytotoxic activity of CD19-specific CAR T cells (Figure 3C, Online Supplementary Figure S2D). Followup showed that the cell product continued to self-deplete from the residual B cells over time (Figure 3C). The presence of CD19+ cells in the manufacturing process may have been the stimulus for the excellent T-cell expansion we observed in the TCR- cells during the manufacturing process.
We next phenotyped the resulting CAR T cells after the editing process and found that the ratio of CD4 to CD8 cells in the resulting CAR T cell product was about 0.8:1 (Figure 3D and F). The majority of CD4+ cells were of naïve and central memory phenotype, while in the CD8+ subpopulation the majority of cells showed a naïve phenotype (Figure 3E and G). This confirms that the CAR T cells had a balanced CD4:CD8 ratio, and that despite the TCR stimulation before genome editing and the transient activation through the CAR mediated by B-cell cytotoxicity, both subpopulations had a high fraction of naïve and CM cells.
Antileukemic efficacy in vivo
We determined the activity of the αβTCR-CD19 CAR T-cell product in vivo using a standard Nalm6 xenograft model.47 We transplanted 5x105 CD19+ Nalm6 cells i.v. into NSG mice to create the CD19+ leukemia model. Four days later, we i.v. infused 1x106 or 5x106 αβTCR-CD19 CAR T or control cells and followed leukemia burden by bioluminescence imaging. The higher dose of CAR T cells led to rapid and complete eradication of leukemia which lasted for at least 3 months, while the lower dose led to a transient decrease in leukemia burden and improved survival although the mice eventually relapsed (Figure 4A). While control mice became moribund and died from disease within 4 weeks, life was significantly extended at both doses of CAR T cells (Figure 4B) (log-rank test: P<0.01 for 5x106 cells, P<0.05 for 1x106 cells). No xenogeneic GvHD was observed in any of the mice, demonstrating the low GvHD potential of the T-cell products. We repeated the experiment at the CAR T-cell dose level of 5x106 cells per mouse using two different control groups, either mock-treated T cells (expressing their endogenous TCR) or RNP-treated T cells (TCR knockout), which confirmed comparable CAR T-cell efficacy and showed no difference between those control groups (Online Supplementary Figure S3A, B).
Off-target evaluation
While the sgRNA specificity has previously been evaluated in an integration-deficient lentivirus capture assay,40 we extended the specificity analysis to measure the off-target activity of the TRAC-targeting RNP using targeted next-generation sequencing. We created a list of predicted off-target sites determined by the COSMID online tool (Figure 5A) and performed targeted deep sequencing of the sites in T cells, from six different donors, electroporated with the RNP (or mock electroporated to determine background). Sequencing confirmed the specificity of the endonuclease with high activity at the on-target site, but no detectable insertions/deletions at off-target sites above the detection limit of 0.1% in any of the samples (Figure 5B).
Discussion
In the case of persistent minimal residual disease after HSCT, relapse risk is high, but treatment options are limited during the time of engraftment and immune recovery. Many immune-based therapies are futile in this period as the immune system is only slowly developing, and transplant protocols typically include immune suppression which would inhibit any adoptive cell-based therapy suchs as CAR T cells. αβhaplo-HSCT represents an excellent platform for adoptive immunotherapy not only because it helps to overcome the limited availability of HLA-matched donors, but also because post-HSCT immunosuppression is not required. It has shown robust clinical results in pediatric patients,8 but some patients still relapse.
We here hypothesize that a donor-derived CAR T-cell product with TCR knockout after haplo-HSCT has the potential to dissociate the beneficial anti-leukemic activity from the harmful GvHD, two phenomena that are inherently connected to each other when infusing unmanipulated donor-derived lymphocytes (donor lymphocyte infusion). This would take advantage of both the graftversus- leukemia effect of allogeneic HSCT and the antileukemic activity of CAR T cells. It will also supplement the polyclonal, HLA-dependent immune response that the transplanted immune system elicits after HSCT with the antigen-specific, HLA-independent cytotoxicity of CAR T cells, in order to address relapses after allogeneic HSCT that occur due to downregulation of HLA molecules. 48-50 Moreover, manufacturing CAR T cells from the donor would maintain immune tolerance between the CAR T-cell product and the donor immune system (which are HLA identical) while taking advantage of the beneficial features of healthy donor T cells.
Our innovative approach avoids the risk of manufacturing failures that comes with the use of autologous T cells, but is distinct from allogeneic, “off-the-shelf” CAR T cells, as it creates a personalized CAR T-cell product for every patient from the respective haploidentical donor. It will therefore not benefit from the same cost-effectiveness that “off-the-shelf” CAR T cells promise, which aim to reduce prices by manufacturing doses for multiple patients during a single run. On the other hand, creating αβTCR-CD19 CAR T cells from the left-over cell fraction and administering them after HSCT will be more economic than the common practice of following the administration of autologous CAR T cells with allogeneic HSCT,51 which carries the high price tag of current CAR T-cell products but then results in their eradication by the donor immune system. Our proposed protocol, in contrast, allows for increased CAR T-cell persistence, since the cells are HLA-identical to the immune system after HSCT. This could create prolonged antileukemic surveillance from a single cell dose, or alternatively enable administration of multiple CAR T-cell doses for the same patient created from one manufacturing run. Moreover, the cells would benefit from the lymphopenia after HSCT, enabling their engraftment and prolonged activity without additional lymphodepleting therapy. Importantly, our approach allows for the creation of both the product containing the hematopoietic stem cells and the gene edited CAR T cells from a single apheresis, as the CAR T cells are made from the otherwise discarded cell fraction. This avoids an additional procedure and thereby leads to cost reduction and mitigates the risks and discomfort for the donor. This will be of particular importance when very young persons are the HSCT donors, e.g., younger siblings of pediatric patients, or the children of adult patients.
Clinical trials have shown that CAR T cells lead to increased survival if disease burden is low before administration of the cells,52 and that high disease burden53 is associated with an increased risk of side effects such as cytokine release syndrome. A potential advantage of using CAR T cells after HSCT is therefore the diminished leukemia burden due to the conditioning regimen and the transplant, which might translate into improved outcomes of the CAR T treatment and a lower incidence of cytokine release syndrome, but in the absence of informative animal models this needs to be tested in a clinical trial.
An alternative haplo-HSCT approach using post-transplant cyclophosphamide for in-vivo T-cell depletion has shown promising results, although published work, to date, has focused primarily on adults.54,55 Although the use of αβhaplo-HSCT requires specific manufacturing expertise and upfront costs to establish the graft processing, we believe that the absence of post-HSCT pharmacological GvHD prophylaxis, the very low rate of severe GvHD and the low infection rate render this approach ideal for combination with post-HSCT adoptive immunotherapy. Eventually, it will need to be determined in prospective trials comparing αβhaplo-HSCT and post-transplant cyclophosphamide in children which alternative is the optimal treatment under what circumstances.
CAR T cells from healthy, allogeneic donors - which have preserved T-cell numbers and functionality and promise to overcome the manufacturing challenges and product variability of autologous CAR T cells29,56-59 – carry the potential to mediate GvHD if they still carry their endogenous TCR.60 Only a limited number of patients have been treated with allogeneic CAR T cells but the frequency of GvHD was surprisingly low when a co-stimulatory domain derived from the CD28 molecule was used in the CAR construct,43 which raises the question of whether TCR deletion is necessary. Mechanistic studies suggest that the simultaneous activation of both the CD28-co-stimulated CAR and the TCR can lead to exhaustion and clonal deletion of alloreactive cells.44 The selective deletion of alloreactive T cells in this mode occurred, however, only at certain ratios between CAR T cells and target cells, and an excess of CAR T cells was able to induce GvHD.44 Furthermore, most CAR T-cell products are transfused without selection of the transduced cells and therefore contain untransduced cells not expressing a CAR which retain their alloreactive potential. Removal of the TCR from the cell surface, e.g., by genome editing approaches,37,38,46 is the best approach to reduce the risk of GvHD of allogeneic cells and additionally might prevent the induction of the T-cell dysfunction that can develop if the CAR and TCR are engaged on the same cell.61 CAR T cells with genome editing-based disruption of the TCR are currently being explored in clinical trials.62
Although CAR T cells with TCR knockout are often referred to as “universal” cells, they can still be rejected by the host immune system as it recovers from the immuno - depletion given prior to CAR T-cell infusion.63 Additional genetic engineering has been proposed to prevent recognition by the host immune system, e.g., the use of genome editing to remove HLA class 1 expression64,65 and the expression of molecules that suppress natural killer cell activity.66 These strategies raise the issue that, if they succeed, cells completely avoid recognition and clearance by the host immune system, but also escape immune surveillance in the case they become infected with viruses or turn malignant. Therefore, engineering an allogeneic CAR Tcell graft that achieves bidirectional immune tolerance with a host immune system including satisfactory immune surveillance remains an unresolved challenge. The use of donor-derived T cells to create a TCR– CAR Tcell product that is administered after allogeneic HSCT enables HLA compatibility of the CAR T cells with the donor-derived host immune system after immune reconstitution. A remaining limitation to full immune compatibility is the nature or the CAR that we used, which is a synthetic protein with non-human parts and potential immunogenicity. Fully humanized CAR are currently in early stages of clinical trials.67 It is possible that in the posttransplant setting the development of an immune response to the CAR will not occur but that can only be tested in a human clinical trial.
It has previously been shown that transfusion of 104 T cells/kg can mediate rapid and protective immune reconstitution, 68 while among a cohort of 98 patients undergoing αβhaplo-HSCT who received a median of 4x104 TCRαβ+ cells, no patient developed high-grade acute GvHD and only one patient developed extensive chronic GvHD.8 With the TCRαβ+ depletion efficiency that we demonstrated, we estimate that therapeutically relevant doses of the cell product (theoretically up to 33x106 cells/kg) could be infused without administering more than 104 TCR+ cells/kg. It remains to be evaluated in a clinical trial whether the small number of αβT cells that are transfused with the hematopoietic stem cell fraction, together with the residual TCR+ cells in the CAR T-cell product, substantially increase the GvHD risk.
In conclusion, we here establish a preclinical proof-ofconcept for using the non-target fraction that is normally discarded during αβ+ T cell/CD19+ B-cell depletion to engineer a CD19-specific CAR T-cell product with a low risk of causing GvHD. αβhaplo-HSCT combined with graft-derived αβTCR-CD19 CAR T cells represents an appealing combination that: (i) allows a donor to be identified for virtually every patient in need; (ii) overcomes the issues related to manufacturing autologous CAR T cells; (iii) abrogates the risk of GvHD through genome editing of the TRAC locus; and (iv) provides persistent targeted immune surveillance after HSCT. Furthermore, the use of adoptive post-HSCT immunotherapy can potentially translate in the future into desirable conditioning regimens with lower toxicity and better preservation of fertility.
Footnotes
- Received August 15, 2019
- Accepted March 19, 2020
Correspondence
Disclosures
Patent submissions are planned. MHP serves on the scientific advisory boards of and has equity in CRISPR Tx and Allogene Therapeutics. Neither companies had any input into the design, execution, interpretation, or publication of this research. All the other authors declare that they have no competing financial conflicts.
Contributions
Conception and design: VW, RB, AB and MHP; in vitro studies: VW and PL; in vivo studies: VW and NM; off-target analysis: CML and GB; data collection: VW, CML; analysis and interpretation of data: VW, MHP and AB; supervision: AB, MHP and MGR; writing of the manuscript: VW and MHP. All authors reviewed the manuscript.
References
- Schrappe M, Hunger SP, Pui C-H. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N Engl J Med. 2012; 366(15):1371-1381. https://doi.org/10.1056/NEJMoa1110169PubMedPubMed CentralGoogle Scholar
- Balduzzi A, Valsecchi MG, Uderzo C. Chemotherapy versus allogeneic transplantation for very-high-risk childhood acute lymphoblastic leukaemia in first complete remission: comparison by genetic randomisation in an international prospective study. Lancet. 2005; 366(9486):635-642. https://doi.org/10.1016/S0140-6736(05)66998-XPubMedGoogle Scholar
- Peters C, Schrappe M, von Stackelberg A. Stem-cell transplantation in children with acute lymphoblastic leukemia: a prospective international multicenter trial comparing sibling donors with matched unrelated donors – the ALL-SCT-BFM-2003 trial. J Clin Oncol. 2015; 33(11):1265-1274. https://doi.org/10.1200/JCO.2014.58.9747PubMedGoogle Scholar
- Horowitz MM, Gale RP, Sondel PM. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990; 75(3):555-562. https://doi.org/10.1182/blood.V75.3.555.555PubMedGoogle Scholar
- Weiden PL, Flournoy N, Thomas ED. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979; 300(19):1068-1073. https://doi.org/10.1056/NEJM197905103001902PubMedGoogle Scholar
- Gragert L, Eapen M, Williams E. HLA match likelihoods for hematopoietic stemcell grafts in the U.S. Registry. N Engl J Med. 2014; 371(4):339-348. https://doi.org/10.1056/NEJMsa1311707PubMedPubMed CentralGoogle Scholar
- Chaleff S, Otto M, Barfield RC. A large-scale method for the selective depletion of alphabeta T lymphocytes from PBSC for allogeneic transplantation. Cytotherapy. 2007; 9(8):746-754. https://doi.org/10.1080/14653240701644000PubMedGoogle Scholar
- Bertaina A, Zecca M, Buldini B. Unrelated donor vs HLA-haploidentical alpha/beta T-cell and B-cell depleted HSCT in children with acute leukemia. Blood. 2018; 132(24):2594-2607. https://doi.org/10.1182/blood-2018-07-861575PubMedGoogle Scholar
- Lang P, Feuchtinger T, Teltschik H-M. Improved immune recovery after transplantation of TCRαβ/CD19-depleted allografts from haploidentical donors in pediatric patients. Bone Marrow Transplant. 2015; 50(S2):S6-S10. https://doi.org/10.1038/bmt.2015.87PubMedGoogle Scholar
- Locatelli F, Merli P, Pagliara D. Outcome of children with acute leukemia given HLA-haploidentical HSCT after αβTcell and B-cell depletion. Blood. 2017; 130(5):677-685. https://doi.org/10.1182/blood-2017-04-779769PubMedGoogle Scholar
- Locatelli F, Bauquet A, Palumbo G, Moretta F, Bertaina A. Negative depletion of /+ T cells and of CD19+ B lymphocytes: a novel frontier to optimize the effect of innate immunity in HLA-mismatched hematopoietic stem cell transplantation. Immunol Lett. 2013; 155(1-2):21-23. https://doi.org/10.1016/j.imlet.2013.09.027PubMedGoogle Scholar
- Locatelli F, Pende D, Mingari MC. Cellular and molecular basis of haploidentical hematopoietic stem cell transplantation in the successful treatment of high-risk leukemias: role of alloreactive NK cells. Front Immunol. 2013; 4:15. https://doi.org/10.3389/fimmu.2013.00015PubMedPubMed CentralGoogle Scholar
- de Witte MA, Kuball J, Miller JS. NK cells and T cells for relapse protection after allogeneic hematopoietic cell transplantation (HCT). Curr Stem Cell Rep. 2017; 3(4):301-311. https://doi.org/10.1007/s40778-017-0106-4PubMedPubMed CentralGoogle Scholar
- Klingebiel T, Cornish J, Labopin M. Results and factors influencing outcome after fully haploidentical hematopoietic stem cell transplantation in children with very high-risk acute lymphoblastic leukemia: impact of center size: an analysis on behalf of the Acute Leukemia and Pediatric Disease Working Parties of the European Blood and Marrow Transplant group. Blood. 2010; 115(17):3437-3446. https://doi.org/10.1182/blood-2009-03-207001PubMedGoogle Scholar
- Kongtim P, Lee DA, Cooper LJN, Kebriaei P, Champlin RE, Ciurea SO. Haploidentical hematopoietic stem cell transplantation as platform for post-transplant cellular therapy. Biol Blood Marrow Transplant. 2015; 21(10):1714. https://doi.org/10.1016/j.bbmt.2015.07.002PubMedPubMed CentralGoogle Scholar
- Lutz C, Massenkeil G, Nagy M. A pilot study of prophylactic donor lymphocyte infusions to prevent relapse in adult acute lymphoblastic leukemias after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2008; 41(9):805-812. https://doi.org/10.1038/sj.bmt.1705981PubMedGoogle Scholar
- Liga M, Triantafyllou E, Tiniakou M. High alloreactivity of low-dose prophylactic donor lymphocyte infusion in patients with acute leukemia undergoing allogeneic hematopoietic cell transplantation with an alemtuzumab-containing conditioning regimen. Biol Blood Marrow Transplant. 2013; 19(1):75-81. https://doi.org/10.1016/j.bbmt.2012.07.021PubMedGoogle Scholar
- Bader P, Klingebiel T, Schaudt A. Prevention of relapse in pediatric patients with acute leukemias and MDS after allogeneic SCT by early immunotherapy initiated on the basis of increasing mixed chimerism: a single center experience of 12 children. Leukemia. 1999; 13(12):2079-2086. https://doi.org/10.1038/sj.leu.2401581PubMedGoogle Scholar
- Dominietto A, Pozzi S, Miglino M. Donor lymphocyte infusions for the treatment of minimal residual disease in acute leukemia. Blood. 2007; 109(11):5063-5064. https://doi.org/10.1182/blood-2007-02-072470PubMedGoogle Scholar
- Lankester AC, Bierings MB, van Wering ER. Preemptive alloimmune intervention in high-risk pediatric acute lymphoblastic leukemia patients guided by minimal residual disease level before stem cell transplantation. Leukemia. 2010; 24(8):1462-1469. https://doi.org/10.1038/leu.2010.133PubMedGoogle Scholar
- Willasch AM, Salzmann-Manrique E, Krenn T. Treatment of relapse after allogeneic stem cell transplantation in children and adolescents with ALL: the Frankfurt experience. Bone Marrow Transplant. 2017; 52(2):201-208. https://doi.org/10.1038/bmt.2016.224PubMedGoogle Scholar
- Anandi P, Tian X, Ito S. Ex vivo T-cell– depleted allogeneic stem cell transplantation for hematologic malignancies: the search for an optimum transplant T-cell dose and T-cell add-back strategy. Cytotherapy. 2017; 19(6):735-743. https://doi.org/10.1016/j.jcyt.2017.03.010PubMedGoogle Scholar
- Di Stasi A, Tey S-K, Dotti G. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011; 365(18):1673-1683. https://doi.org/10.1056/NEJMoa1106152PubMedPubMed CentralGoogle Scholar
- Bonini C, Ferrari G, Verzeletti S. HSVTK gene transfer into donor lymphocytes for control of allogeneic graft-versusleukemia. Science. 1997; 276(5319):1719-1724. https://doi.org/10.1126/science.276.5319.1719PubMedGoogle Scholar
- Locatelli F, Ruggeri A, Merli P. Administration of BPX-501 cells following AT and B-cell-depleted HLA haploidentical HSCT (haplo-HSCT) in children with acute leukemias. Blood. 2018; 132(Suppl 1):307. https://doi.org/10.1182/blood-2018-99-119481PubMedGoogle Scholar
- Majzner RG, Heitzeneder S, Mackall CL. Harnessing the immunotherapy revolution for the treatment of childhood cancers. Cancer Cell. 2017; 31(4):476-485. https://doi.org/10.1016/j.ccell.2017.03.002PubMedGoogle Scholar
- Rivière I, Sadelain M. Chimeric antigen receptors: a cell and gene therapy perspective. Mol Ther. 2017; 25(5):1117-1124. https://doi.org/10.1016/j.ymthe.2017.03.034PubMedPubMed CentralGoogle Scholar
- Sadelain M. CD19 CAR T cells. Cell. 2017; 171(7):1471. https://doi.org/10.1016/j.cell.2017.12.002PubMedGoogle Scholar
- Ruella M, Xu J, Barrett DM. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nat Med. 2018; 24(10):1499-1503. https://doi.org/10.1038/s41591-018-0201-9PubMedPubMed CentralGoogle Scholar
- Fraietta JA, Nobles CL, Sammons MA. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018; 558(7709):307-312. https://doi.org/10.1038/s41586-018-0178-zPubMedPubMed CentralGoogle Scholar
- Nicholson IC, Lenton KA, Little DJ. Construction and characterisation of a functional CD19 specific single chain Fv fragment for immunotherapy of B lineage leukaemia and lymphoma. Mol Immunol. 1997; 34(16-17):1157-1165. https://doi.org/10.1016/S0161-5890(97)00144-2PubMedGoogle Scholar
- Bak RO, Dever DP, Porteus MH. CRISPR/Cas9 genome editing in human hematopoietic stem cells. Nat Protoc. 2018; 13(2):358-376. https://doi.org/10.1038/nprot.2017.143PubMedPubMed CentralGoogle Scholar
- Haeussler M, Schönig K, Eckert H. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biol. 2016; 17(1):148. https://doi.org/10.1186/s13059-016-1012-2PubMedPubMed CentralGoogle Scholar
- Cradick TJ, Qiu P, Lee CM, Fine EJ, Bao G. COSMID: a web-based tool for identifying and validating CRISPR/Cas off-target sites. Mol Ther Nucleic Acids. 2014; 3:e214. https://doi.org/10.1038/mtna.2014.64PubMedPubMed CentralGoogle Scholar
- Lee CM, Cradick TJ, Bao G. The Neisseria meningitidis CRISPR-Cas9 system enables specific genome editing in mammalian cells. Mol Ther. 2016; 24(3):645-654. https://doi.org/10.1038/mt.2016.8PubMedPubMed CentralGoogle Scholar
- MacLeod DT, Antony J, Martin AJ. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells. Mol Ther. 2017; 25(4):949-961. https://doi.org/10.1016/j.ymthe.2017.02.005PubMedPubMed CentralGoogle Scholar
- Eyquem J, Mansilla-Soto J, Giavridis T. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017; 543(7643):113-117. https://doi.org/10.1038/nature21405PubMedPubMed CentralGoogle Scholar
- Provasi E, Genovese P, Lombardo A. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat Med. 2012; 18(5):807-815. https://doi.org/10.1038/nm.2700PubMedPubMed CentralGoogle Scholar
- Long AH, Haso WM, Shern JF. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015; 21(6):581-590. https://doi.org/10.1038/nm.3838PubMedPubMed CentralGoogle Scholar
- Osborn MJ, Webber BR, Knipping F. Evaluation of TCR gene editing achieved by TALENs, CRISPR/Cas9, and megaTAL nucleases. Mol Ther. 2016; 24(3):570-581. https://doi.org/10.1038/mt.2015.197PubMedPubMed CentralGoogle Scholar
- Vakulskas CA, Dever DP, Rettig GR. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat Med. 2018; 24(8):1216-1224. https://doi.org/10.1038/s41591-018-0137-0PubMedPubMed CentralGoogle Scholar
- Hendel A, Bak RO, Clark JT. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nat Biotechnol. 2015; 33(9):985-989. https://doi.org/10.1038/nbt.3290PubMedPubMed CentralGoogle Scholar
- Qasim W. Allogeneic CAR T cell therapies for leukemia. Am J Hematol. 2019; 94(S1):S50-S54. https://doi.org/10.1002/ajh.25399PubMedGoogle Scholar
- Ghosh A, Smith M, James SE. Donor CD19 CAR T cells exert potent graft-versuslymphoma activity with diminished graftversus- host activity. Nat Med. 2017; 23(2):242-249. https://doi.org/10.1038/nm.4258PubMedPubMed CentralGoogle Scholar
- Poirot L, Philip B, Schiffer-Mannioui C. Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive Tcell immunotherapies. Cancer Res. 2015; 75(18):3853-3864. https://doi.org/10.1158/0008-5472.CAN-14-3321PubMedGoogle Scholar
- Qasim W, Zhan H, Samarasinghe S. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med. 2017; 9(374)https://doi.org/10.1126/scitranslmed.aaj2013PubMedGoogle Scholar
- Barrett DM, Seif AE, Carpenito C. Noninvasive bioluminescent imaging of primary patient acute lymphoblastic leukemia: a strategy for preclinical modeling. Blood. 2011; 118(15):e112-e117. https://doi.org/10.1182/blood-2011-04-346528PubMedPubMed CentralGoogle Scholar
- Sano H, Mochizuki K, Kobayashi S. Two occurrences of leukemia relapse due to mismatched HLA loss after haploidentical stem cell transplantation from different family donors with KIR ligand mismatch. J Pediatr Hematol Oncol. 2020; 42(2):e104-e106. https://doi.org/10.1097/MPH.0000000000001443PubMedGoogle Scholar
- Christopher MJ, Petti AA, Rettig MP. Immune escape of relapsed AML cells after allogeneic transplantation. N Engl J Med. 2018; 379(24):2330-2341. https://doi.org/10.1056/NEJMoa1808777PubMedPubMed CentralGoogle Scholar
- Vago L, Perna SK, Zanussi M. Loss of mismatched HLA in leukemia after stem-cell transplantation. N Engl J Med. 2009; 361(5):478-488. https://doi.org/10.1056/NEJMoa0811036PubMedGoogle Scholar
- Kenderian SS, Porter DL, Gill S. Chimeric antigen receptor T cells and hematopoietic cell transplantation: how not to put the CART before the horse. Biol Blood Marrow Transplant. 2017; 23(2):235-246. https://doi.org/10.1016/j.bbmt.2016.09.002PubMedPubMed CentralGoogle Scholar
- Park JH, Rivière I, Gonen M. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018; 378(5):449-459. https://doi.org/10.1056/NEJMoa1709919PubMedPubMed CentralGoogle Scholar
- Maude SL, Frey N, Shaw PA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371(16):1507-1517. https://doi.org/10.1056/NEJMoa1407222PubMedPubMed CentralGoogle Scholar
- Luznik L, Fuchs EJ. High-dose, post-transplantation cyclophosphamide to promote graft-host tolerance after allogeneic hematopoietic stem cell transplantation. Immunol Res. 2010; 47(1-3):65-77. https://doi.org/10.1007/s12026-009-8139-0PubMedPubMed CentralGoogle Scholar
- Luznik L, Bolaños-Meade J, Zahurak M. High-dose cyclophosphamide as singleagent, short-course prophylaxis of graft-versus- host disease. Blood. 2010; 115(16):3224-3230. https://doi.org/10.1182/blood-2009-11-251595PubMedPubMed CentralGoogle Scholar
- Hoffmann J-M, Schubert M-L, Wang L. Differences in expansion potential of naive chimeric antigen receptor T cells from healthy donors and untreated chronic lymphocytic leukemia patients. Front Immunol. 2018; 8:1956. https://doi.org/10.3389/fimmu.2017.01956PubMedPubMed CentralGoogle Scholar
- Chongsathidkiet P, Jackson C, Koyama S. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat Med. 2018; 24(9):1459-1468. https://doi.org/10.1038/s41591-018-0135-2PubMedPubMed CentralGoogle Scholar
- Qin H, Ishii K, Nguyen S. Murine Pre- B cell ALL induces T cell dysfunction not fully reversed by introduction of a chimeric antigen receptor. Blood. 2018; 132(18):1899-1910. https://doi.org/10.1182/blood-2017-12-815548PubMedPubMed CentralGoogle Scholar
- Knaus HA, Berglund S, Hackl H. Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight. 2018; 3(21)https://doi.org/10.1172/jci.insight.120974PubMedPubMed CentralGoogle Scholar
- Dai H, Zhang W, Li X. Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology. 2015; 4(11):e1027469. https://doi.org/10.1080/2162402X.2015.1027469PubMedPubMed CentralGoogle Scholar
- Yang Y, Kohler ME, Chien CD. TCR engagement negatively affects CD8 but not CD4 CAR T cell expansion and leukemic clearance. Sci Transl Med. 2017; 9(417)https://doi.org/10.1126/scitranslmed.aag1209PubMedPubMed CentralGoogle Scholar
- Zhao J, Song Y, Liu D. Clinical trials of dualtarget CAR T cells, donor-derived CAR T cells, and universal CAR T cells for acute lymphoid leukemia. J Hematol Oncol. 2019; 12(1):17. https://doi.org/10.1186/s13045-019-0705-xPubMedPubMed CentralGoogle Scholar
- Torikai H, Reik A, Liu P-Q. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19- specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012; 119(24):5697-5705. https://doi.org/10.1182/blood-2012-01-405365PubMedPubMed CentralGoogle Scholar
- Torikai H, Reik A, Soldner F. Toward eliminating HLA class I expression to generate universal cells from allogeneic donors. Blood. 2013; 122(8):1341-1349. https://doi.org/10.1182/blood-2013-03-478255PubMedPubMed CentralGoogle Scholar
- Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res. 2017; 23(9):2255-2266. https://doi.org/10.1158/1078-0432.CCR-16-1300PubMedPubMed CentralGoogle Scholar
- Gornalusse GG, Hirata RK, Funk SE. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat Biotechnol. 2017; 35(8):765-772. https://doi.org/10.1038/nbt.3860PubMedPubMed CentralGoogle Scholar
- Brudno JN, Lam N, Vanasse D. Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma. Nat Med. 2020; 26(2):270-280. https://doi.org/10.1038/s41591-019-0737-3PubMedPubMed CentralGoogle Scholar
- Zhou X, Dotti G, Krance RA. Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation. Blood. 2015; 125(26):4103-4113. https://doi.org/10.1182/blood-2015-02-628354PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.