Expansion of Donor NK Cells for Adoptive Immunotherapy in Haploidentical Stem Cell Transplantation: A Phase I–II Study

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3893-3893
Author(s):  
Uwe Siegler ◽  
Sandrine Meyer-Monard ◽  
Simon Jörger ◽  
Martin Stern ◽  
Andre Tichelli ◽  
...  

Abstract Natural killer (NK) lymphocytes are essential for anti-cancer defense. Transplantations of haploidentical hematopoietic stem cells (HSCT) across HLA class I barriers highlighted the graft-versus-leukemia effect of alloreactive NK cells and strengthened prospects for NK cell immunotherapy in human cancer. Reconstituting NK cells remain immature with impaired cytotoxicity for more than 6 months after HSCT, suggesting a benefit of adoptive transfer of mature NK cells to provide the patient with competent cytotoxic effectors. We previously reported the feasibility of purifying NK cells in numbers sufficient for infusions of 1×107 cells/kg (Passweg et al. 2004). Here we describe an approach of increasing the effector to target ratio by large-scale good manufacturing practice (GMP)-compliant expansion of NK cells and preemptive multiple donor lymphocyte infusions (NK-DLI) after haploidentical HSCT for hematological malignancies. This single centre phase I–II clinical study was approved by the ethical committee. NK cells were purified from a median of 7.4L (range: 5.6–10.0L) unstimulated leukapheresis of 5 healthy donors by T cell depletion and subsequent NK cell selection with anti-CD3 and anti-CD56 coated immunomagnetic microbeads on the CliniMACS_ device. A median of 5.32×108 (2.87–6.42×108) CD56+CD3- NK cells was obtained with a purity of 98.5% (71.9–99.6%), and a residual CD3+ T cell content of 0.030% (<0.001–0.070%) corresponding to a T cell depletion efficiency of 3.51 log (3.15–4.87 log). NK cells were cultured in 30 air-permeable bags and up to 7.8L of GMP-certified medium containing human serum, IL-2, anti-CD3 monoclonal antibody OKT-3 and irradiated autologous feeder cells. After 15–20 days of culture, NK cell numbers increased on average 50 fold (12–137 fold). CD3+ T cells concomitantly expanded to 0.59% (0.014–1.68%) of total cells. A second CD3 depletion was performed with 3 NK cell products which exceeded the maximal T cell dose of 0.5×105/kg as required by the planned therapeutic study protocol. T cell content was reduced by 1.07 log (0.84–3.17 log) at the expense of a significant NK cell loss of 47% (32–76%). Bacterial contamination tests from all culture bags were negative. These results demonstrate the feasibility of clinical grade large-scale ex vivo NK cell expansion. GMP-expanded NK cells exhibited high levels of activating receptors NKG2D and NKp44 (3.1 and 16.8 fold increase above basal levels). The proportions of alloreactive NK cells with single killer immunoglobulin-like receptor (KIR) specificities remained stable comprising 2.4–16.0% and 5.2–14.2% of cells before and after expansion. GMP-expanded NK cells were more cytotoxic against K562 target cells than freshly isolated NK cells (mean±SEM: 61.0±3.1% vs. 27.0±8.0% specific lysis at 10:1 effector to target ratio). The cytolytic activity against KIR-HLA class I mismatched primary AML blasts was 7.0±3.7%, reflecting the content of alloreactive NK cell subsets. This is supported by our finding that purified single-KIR alloreactive NK cells lysed mismatched AML blasts with a high efficiency (22.0±6.3% at 10:1 ratio), indicating the anti-leukemic capacity of GMP-expanded NK cells. Two NK-DLI products with a total of 21.0×108 and 189.4×108 NK cells were generated for two patients and divided into three escalating doses (1×106, 1×107 and 1.5×107/kg) for patient 1 and into four doses (1×106, 1×107 and twice 1×108/kg) for patient 2. The infusions were tolerated without any acute adverse effects. In conclusion, we established and clinically implemented a protocol suitable for GMPcompliant expansion of cytokine-activated NK cells allowing multiple infusions of high numbers of cells with strong cytolytic activity. These results provide the basis for a prospective clinical efficacy trial to advance the therapeutic field of NK-DLI against human cancer.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3002-3002 ◽  
Author(s):  
Daniela Pende ◽  
Stefania Marcenaro ◽  
Michela Falco ◽  
Stefania Martini ◽  
Maria Ester Bernardo ◽  
...  

Abstract T-cell depleted hematopoietic stem cell transplantation from haploidentical donors (haplo-HSCT) has been reported to benefit from the graft-versus-leukemia effect mediated by natural killer (NK) cells when donor displays NK alloreactivity versus the recipient. NK alloreactivity is mediated by NK receptors, namely Killer Ig-like receptors (KIR) which are specific for allotypic determinants that are shared by different HLA-class I alleles (referred to as KIR ligands). It is known that KIR2DL1 recognizes HLA-C alleles characterized by Lys at position 80 (C2 group), KIR2DL2/3 recognize HLA-C alleles characterized by Asn at position 80 (C1 group), KIR3DL1 recognizes HLA-B alleles sharing the Bw4 supertypic specificity (Bw4 group) and KIR3DL2 recognizes HLA-A3 and –A11 alleles. KIR2D/3DL are inhibitory receptors that, upon engagement with the cognate ligand, inhibit lysis. Activating KIRs, highly homologous in the extracellular domain to the inhibitory counterparts, are KIR2DS1, KIR2DS2 and KIR3DS1, but only KIR2DS1 has been shown to specifically recognize C2 group of alleles expressed on B-EBV cells. We analyzed 21 children with leukemia receiving haplo-HSCT from a relative after a myeloablative conditioning regimen; in all pairs, the expression of a given KIR ligand (HLA class I allele) of the donor was missing in the patient (i.e. KIR ligand-mismatched haplo-HSCT). T-cell depletion was performed through positive selection of CD34+ cells; no pharmacological immune suppression was employed after HSCT. KIR genotype of all donors was evaluated to detect the presence of the various inhibitory and activating KIR genes. Phenotypic analyses were performed on NK cells derived from the donor and the patient at different time points after HSCT. Thanks to the availability of new mAbs able to discriminate between the inhibitory and the activating forms of a certain KIR, we could identify the alloreactive NK cell subset at the population level. These alloreactive NK cells express the KIR specific for the KIR ligand-mismatch (permissive inhibitory KIR) and the activating KIR (if present), while they do not express all inhibitory KIR specific for the patient HLA alleles and NKG2A. Thus, in most instances, we could precisely identify the size of the alloreactive NK cell subset in the donor and in the reconstituted repertoire of the recipient. Functional assays were performed to assess alloreactivity, using appropriate B-EBV cell lines and, if available, patient’s leukemia blasts. In some cases, also NK cell clones were extensively studied, for phenotype and receptor involvement in killing activity. We found that, in most transplanted patients, variable proportions of donor-derived alloreactive NK cells displaying anti-leukemia activity were generated and maintained even at late time-points after transplantation. Donor-derived KIR2DL1+ NK cells isolated from the recipient displayed the expected capability of selectively killing C1/C1 target cells, including patient leukemia blasts. Differently, KIR2DL2/3+ NK cells displayed poor alloreactivity against leukemia cells carrying HLA alleles belonging to the C2 specificity. Unexpectedly, this was due to recognition of C2 by KIR2DL2/3, as revealed by receptor blocking experiments and by binding assays of soluble KIR to HLA-C transfectants. Remarkably, however, C2/C2 leukemia blasts were killed by KIR2DL2/3+ (or by NKG2A+) NK cells that co-expressed KIR2DS1. This could be explained by the ability of KIR2DS1 to directly recognize C2 on leukemia cells. A role for the KIR2DS2 activating receptor in leukemia cell lysis could not be established. Taken together, these findings provide new information on NK alloreactivity in haplo-HSCT that may greatly impact on the selection of the optimal donor.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1838-1838 ◽  
Author(s):  
Yvonne A. Efebera ◽  
Ashley E Rosko ◽  
Craig Hofmeister ◽  
Joe Benner ◽  
Courtney Bakan ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is associated with profound and widespread disarray of both the adaptive and innate arms of the immune system including loss of effector T cell function, humoral immune deficiency, and natural killer (NK) cell immunity. This immunosuppressive milieu is crucial to promoting disease progression. Standard treatment options (immunomodulators (IMIDs) and proteosome inhibitors, radiation, and high-dose corticosteroids) offer modest benefit, but also contribute to further immune suppression. Little is known regarding the mechanisms by which immune dysfunction and immunoevasion occur. Our group has characterized an important role for the programmed death receptor-1 (PD-1) / PD-L1 signaling axis in these processes. MDV9300 (formerly CT-011 / Pidilizumab) is a novel IgG1 humanized monoclonal antibody (mAb) that modulates the immune response through interaction with PD-1. Lenalidomide (Len) an IMID exerts efficacy in MM in part through enhancement of NK cell versus MM effect - an effect likely mediated through T cell production of interleukin (IL)-2. In our in-vitro study, pretreatment of NK cells with MDV9300 with or without Len enhanced immune complex formation between NK cells and MM tumor targets and also augmented NK cell activation and cytotoxicity against MM. We sought to determine the safety, tolerability and any early signs of efficacy in relapsed or refractory MM patients using MDV9300 in combination with Len. Methods: In the phase I portion, the primary endpoint is to determine the maximum tolerated dose (MTD) of the combination. Key eligibility criteria are relapsed or refractory disease but not progressed on Len 25 mg; ≥2 prior lines of therapy, absolute neutrophil count ≥ 1000/µL; Platelets ≥60,000/µL; and creatinine clearance of ≥ 40ml/min. Patients are treated with escalating doses of MDV9300 and Len utilizing a 3x3 escalation design (Table 1). If stable disease is the best response after 4 cycles, patients have the option of adding dexamethasone (20-40mg weekly). Len dose may be modified independently of MDV9300. Patients can receive a maximum of 12 cycles of therapy. Results: Twelve patients are evaluable to date. The median age was 68.5 (range 49-82) and the median time from diagnosis 4.98 years (range 1.54-12.62). At study entry, 67% had high risk cytogenetics (del 17p, complex karyotype, gain 1q) and the median number of prior treatment lines was 2 (range 2-11). 100% of patients had received prior Len, bortezomib and Dex, 50% alkylating agents (cyclophosphamide, oral melphalan, bendamustine), 75% autologous stem cell transplant, 25% pomalidomide and 33% carfilzomib. MDV9300 infusion has been well tolerated with only one grade 2 infusion related toxicity with sore throat. The patient received hydrocortisone with no further reaction observed. Grade 3/4 Anemia, neutropenia, and thrombocytopenia attributable to therapy have been seen in 25%, 23%, and 34% of patients, respectively. Other common grade 2-3 therapy related adverse events are fatigue (50%), anorexia (17%), and hypophosphatemia (17%). There has been no grade 3 or higher infection and no worsening of neuropathy from baseline. Len dose was reduced in 3 patients (25%) and increased in one. There has been no dose reduction in MDV9300. Dex 20 mg or less was added in 2 patients for muscle cramps and < PR after 3 cycles. To date 7 patients are off therapy; 1 due to grade 3 fatigue and 6 due to disease progression. Five patients continue on therapy at respective 12, 11, 9, 5 and 3 months. Responses to date have been 3 Very good partial response,1 partial response, 2 minimal response and 2 stable disease. Conclusion: The combination of steroid sparing MDV9300 and Len regimen has demonstrated an acceptable toxicity profile to date with evidence of anti-myeloma activity. This is the first reported combination anti-PD-1 based immune therapy for MM. Updated results will be presented at the meeting including the MTD dose for phase II. Table 1. MDV9300- mg/kg Intravenously given on day 3 every 28 days Lenalidomide- mg orally days 1-21 every 28 days DLT Evaluable DLTs Cohort 1 1.5 15 6 Grade 3 fatigue. Cohort extended to 6 Cohort 2 3 15 3 none Cohort 3 3 25 3 none Cohort 4 6 25 0 Acknowledgments: Drug has been provided by Medivation; The study is sponsored by the American Cancer Society Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4718-4718
Author(s):  
Giulia Giunti ◽  
David Malone ◽  
Lucas Chan ◽  
Darling David ◽  
Shahram Y Kordasti ◽  
...  

Abstract Abstract 4718 Improved experimental therapies are needed for Multiple Myeloma (MM). Despite major progress in treatment and initial induction of remission, myeloma remains an incurable disease. Although immunotherapy and, in particular, the employment of NK cells offers an approach of interest for the treatment of Multiple Myeloma (MM), recent studies have shown that myeloma cells utilise a number different mechanisms to impair NK and T cell functions. Important amongst these mechanisms is the reduced expression of CD80 in the sub-populations of PBMC isolated from myeloma patients. We have previously demonstrated CD80/IL-2 mediated stimulation of NK and T cells isolated from AML patients (as measured by proliferation, cytokine release and target cell specific cytolytic activity). In the present study we have examined the ability of genetically modified MM cells engineered to express CD80 and IL-2 to stimulate NK cell functions. These studies confirm the ability of MM cells to suppress NK cell functions in healthy PBMC and show that in contrast to the unmodified MM cells, the CD80/IL-2 expressing MM cells are able to stimulate a moderate increase in NK and T cell numbers and a significant increase in the fraction of NK cells with activatory receptors (NKp44, NKp30, NKp46) and activation markers (CD69) on the cell surface of both NK and T cells. More importantly for potential therapeutic applications the stimulated NK cells show increased cytolytic activity against the unmodified MM cells. This data suggest that CD80/IL-2 MM cells may be able to overcome the immune suppressive functions of unmodified MM cells and to stimulate NK, and T cell mediated responses against the unmodified MM cells. Therefore CD80/IL-2 expressing MM cells may provide a suitable cellular vaccine for NK cell stimulation and possibly the induction of broader ranging immunological responses against multiple myeloma cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 115 (13) ◽  
pp. 2686-2694 ◽  
Author(s):  
Andreas T. Björklund ◽  
Marie Schaffer ◽  
Cyril Fauriat ◽  
Olle Ringdén ◽  
Mats Remberger ◽  
...  

Abstract Natural killer (NK)–cell alloreactivity in recipients of hematopoietic stem cell grafts from HLA-identical siblings is intriguing and has suggested breaking of NK-cell tolerance during the posttransplantation period. To examine this possibility, we analyzed clinical outcomes in a cohort of 105 patients with myeloid malignancies who received T cell–replete grafts from HLA-matched sibling donors. Presence of inhibitory killer cell immunoglobulin-like receptors (KIRs) for nonself HLA class I ligands had no effect on disease-free survival, incidence of relapse, or graft-versus-host disease. A longitudinal analysis of the NK-cell repertoire and function revealed a global hyporesponsiveness of NK cells early after transplantation. Functional responses recovered at approximately 6 months after transplantation. Importantly, NKG2A− NK cells expressing KIRs for nonself HLA class I ligands remained tolerant at all time points. Furthermore, a direct comparison of NK-cell reconstitution in T cell–replete and T cell–depleted HLA-matched sibling stem cell transplantation (SCT) revealed that NKG2A+ NK cells dominated the functional repertoire early after transplantation, with intact tolerance of NKG2A− NK cells expressing KIRs for nonself ligands in both settings. Our results provide evidence against the emergence of alloreactive NK cells in HLA-identical allogeneic SCT.


Blood ◽  
2011 ◽  
Vol 117 (16) ◽  
pp. 4284-4292 ◽  
Author(s):  
Simona Sivori ◽  
Simona Carlomagno ◽  
Michela Falco ◽  
Elisa Romeo ◽  
Lorenzo Moretta ◽  
...  

Abstract In allogeneic HSCT, NK-cell alloreactivity is determined by the presence in the donor of NK cells expressing inhibitory killer cell Ig-like receptors (KIRs) that recognize HLA class I allotypes present in the donor but lacking in the recipient. Dominant KIR ligands are the C1 and C2 epitopes of HLA-C. All HLA-C allotypes have either the C1 epitope, the ligand for KIR2DL2/L3, or the C2 epitope, the ligand for KIR2DL1/S1. Here, we show that, in alloreactive NK-cell responses, KIR2DS1 expression represents a remarkable advantage as it allows efficient killing of C2/C2 or C1/C2 myelomonocitic dendritic cells (DCs) and T-cell blasts. When DCs or T-cell blasts were derived from C2/C2, Bw4/Bw4 donors, the activating signals delivered by KIR2DS1 could override the inhibition generated by NKG2A or KIR2DL2/L3 expressed on the same NK-cell clone. Furthermore, substantial lysis of C2/C2, Bw4/Bw6 targets was mediated by KIR2DS1+ NK cells coexpressing KIR3DL1. Importantly, in the case of C1/C2 targets, KIR2DS1+ NK cells were inhibited by the coexpression of KIR2DL2/L3 but not of NKG2A. Thus, KIR2DS1 expression in HSC donors may substantially increase the size of the alloreactive NK-cell subset leading to an enhanced ability to limit GVHD and improve engrafment.


Blood ◽  
2004 ◽  
Vol 103 (8) ◽  
pp. 3122-3130 ◽  
Author(s):  
Christian Demanet ◽  
Arend Mulder ◽  
Veronique Deneys ◽  
Maria J. Worsham ◽  
Piet Maes ◽  
...  

Abstract Human leukocyte antigen (HLA) class I antigen defects may have a negative impact on the growing application of T-cell–based immunotherapeutic strategies for treatment of leukemia. Therefore in the present study, taking advantage of a large panel of HLA class I allele–specific human monoclonal antibodies, we have compared HLA class I antigen expression on leukemic cells with that on autologous and allogeneic normal cells. Down-regulation of HLA-A and/or -B allospecificities was present in the majority of the patients studied. However, down-regulation did not affect all HLA class I alleles uniformly, but was almost exclusively restricted to HLA-A allospecificities and to HLA-B allospecificities which belong to the HLA-Bw6 group. The latter allospecificities, at variance from those that belong to the HLA-Bw4 group, do not modulate the interactions of leukemic cells with natural killer (NK) cells. Therefore, our results suggest that the selective down-regulation of HLA-A and HLA-Bw6 allospecificities associated with HLA-Bw4 preservation provides leukemic cells with an escape mechanism not only from cytotoxic T lymphocytes (CTLs), but also from NK cells. As a result T-cell–based immunotherapeutic strategies for leukemia should utilize HLA-Bw4 alloantigens as restricting elements since a selective HLA-Bw4 allele loss would provide leukemic cells with an escape mechanism from CTLs, but would increase their susceptibility to NK cell–mediated lysis. (Blood. 2004;103:3122-3130)


2021 ◽  
Vol 11 ◽  
Author(s):  
Keven Hoerster ◽  
Markus Uhrberg ◽  
Constanze Wiek ◽  
Peter A. Horn ◽  
Helmut Hanenberg ◽  
...  

Cellular immunotherapy using chimeric antigen receptors (CARs) so far has almost exclusively used autologous peripheral blood-derived T cells as immune effector cells. However, harvesting sufficient numbers of T cells is often challenging in heavily pre-treated patients with malignancies and perturbed hematopoiesis and perturbed hematopoiesis. Also, such a CAR product will always be specific for the individual patient. In contrast, NK cell infusions can be performed in non-HLA-matched settings due to the absence of alloreactivity of these innate immune cells. Still, the infused NK cells are subject to recognition and rejection by the patient’s immune system, thereby limiting their life-span in vivo and undermining the possibility for multiple infusions. Here, we designed genome editing and advanced lentiviral transduction protocols to render primary human NK cells unsusceptible/resistant to an allogeneic response by the recipient’s CD8+ T cells. After knocking-out surface expression of HLA class I molecules by targeting the B2M gene via CRISPR/Cas9, we also co-expressed a single-chain HLA-E molecule, thereby preventing NK cell fratricide of B2M-knockout (KO) cells via “missing self”-induced lysis. Importantly, these genetically engineered NK cells were functionally indistinguishable from their unmodified counterparts with regard to their phenotype and their natural cytotoxicity towards different AML cell lines. In co-culture assays, B2M-KO NK cells neither induced immune responses of allogeneic T cells nor re-activated allogeneic T cells which had been expanded/primed using irradiated PBMNCs of the respective NK cell donor. Our study demonstrates the feasibility of genome editing in primary allogeneic NK cells to diminish their recognition and killing by mismatched T cells and is an important prerequisite for using non-HLA-matched primary human NK cells as readily available, “off-the-shelf” immune effectors for a variety of immunotherapy indications in human cancer.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A405-A405
Author(s):  
David Hong ◽  
Sandip Patel ◽  
Manish Patel ◽  
Kimberly Musni ◽  
Marlisa Anderson ◽  
...  

BackgroundFT500 is an investigational, off-the-shelf NK cell cancer immunotherapy derived from a human clonal master iPSC line, a renewable cell source from which innate effector cells can be mass produced and made available off-the-shelf for broad patient access and multiple dose administration. FT500 has potent innate cellular cytotoxicity as compared to NK cells sourced from healthy donors and has been shown to synergize with T cells and anti-PD-1 blockade in preclinical studies.1MethodsFT500 is being investigated in a Phase I clinical trial as monotherapy and in combination with immune checkpoint inhibitors (ICIs) in patients with advanced solid tumors and lymphomas (ClinicalTrials.gov: NCT03841110). Treatment consists of 2 days lympho conditioning (fludarabine 25 mg/m2 and cyclophosphamide 300 mg/m2) followed by 2 cycles of 3 once weekly doses of FT500 as monotherapy or combined with 1 of 3 approved ICIs (nivolumab, pembrolizumab, or atezolizumab) in patients who have failed prior ICI therapy. Key clinical and translational readouts include FT500 safety and tolerability, including immune mediated toxicities and anti-product immunogenicity.Results15 patients with relapsed/refractory disease following a median of 4 prior therapies were treated in dose escalation, including 9 with FT500 monotherapy (3 with 1×108 cells, 6 with 3×108 cells) and 6 with FT500 (3 each with 1×108 and 3×108 cells) combined with ICI. No dose limiting toxicities, Grade ³3 related adverse events (AEs), Grade ³3 related serious AEs, or related AEs leading to treatment discontinuation were reported. No graft-versus-host disease (GvHD), cytokine release syndrome (CRS), or neurotoxicity (NT) was observed. The most common treatment-emergent AEs in >3 patients were nausea (9), fatigue (7), constipation, decreased appetite, decreased lymphocyte count, decreased white blood cell count (5 each), anemia, and decreased neutrophil count (4 each). Nine of 13 efficacy-evaluable solid tumor patients had best response of stable disease by iRECIST. One patient with classical Hodgkin lymphoma (cHL) refractory to prior experimental anti-PD-1 therapy had a 58% reduction in target lesions size following FT500 plus ICI. No evidence of robust B- or T-cell mediated anti product responses was observed despite endogenous immune cell recovery following lympho-conditioning.ConclusionsAdministration of 6 doses of up to 3×108 FT500 cells is safe and tolerable without evidence of GvHD, CRS, NT, or host immune rejection. Enrollment of advanced non-small cell lung cancer and cHL patients at 3×108 FT500 cells per dose combined with ICI is ongoing.Ethics ApprovalThis study is being conducted in accordance with the Declaration of Helsinki and was approved by all Institutional Review Boards from each clinical site participating in the study. Specific approval numbers can be provided upon request.ReferenceCichocki F, Bjordahl R, Gaidarova S, Mahmood S, Rogers P, Ge MQ, Kaufman DS, Cooley S, Valamehr B, Miller JS. iPSC-derived NK cells and anti-PD-1 antibody synergize to enhance T cell cytokine and cytolytic responses against multiple tumors. Blood 2018;132(Supplement 1):730.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1711-1711
Author(s):  
Yong Zhang ◽  
Surbhi Goel ◽  
Aaron Prodeus ◽  
Utsav Jetley ◽  
Yiyang Tan ◽  
...  
Keyword(s):  
T Cells ◽  
T Cell ◽  
Nk Cells ◽  
Nk Cell ◽  
Class Ii ◽  

Abstract Introduction. Despite the success of autologous chimeric antigen receptor (CAR)-T cells, barriers to a more widespread use of this potentially curative therapy include manufacturing failures and the high cost of individualized production. There is a strong desire for an immediately available cell therapy option; however, development of "off-the-shelf" T cells is challenging. Alloreactive T cells from unrelated donors can cause graft versus host disease (GvHD) for which researchers have successfully used nucleases to reduce expression of the endogenous T cell receptor (TCR) in the allogeneic product. The recognition of allogeneic cells by the host is a complex issue that has not been fully solved to date. Some approaches utilize prolonged immune suppression to avoid immune rejection and increase persistence. Although showing responses in the clinic, this approach carries the risk of infections and the durability of the adoptive T cells is uncertain. Other strategies include deletion of the B2M gene to remove HLA class I molecules and avoid recognition by host CD8 T cells. However, loss of HLA class I sends a "missing-self" signal to natural killer (NK) cells, which readily eliminate B2Mnull T cells. To overcome this, researchers are exploring insertion of the non-polymorphic HLA-E gene, which can provide partial but not full protection from NK cell-mediated lysis. Because activated T cells upregulate HLA class II, rejection by alloreactive CD4 T cells should also be addressed. Methods. Here, we developed an immunologically stealth "off-the-shelf" T cell strategy by leveraging our CRISPR/Cas9 platform and proprietary sequential editing process. To solve the issue of rejection by alloreactive CD4 and CD8 T cells, we knocked out (KO) select HLA class I and class II expression with a sequential editing process. Additionally, we utilize potent TCR-α and -β constant chain (TRAC, TRBC) gRNAs that achieve &gt;99% KO of the endogenous TCR, addressing the risk of GvHD. An AAV-mediated insertion of a CAR or TCR into the TRAC locus is used in parallel with the TRAC KO step to redirect the T cells to tumor targets of interest. Alloreactivity by CD4 and CD8 T cells, NK killing, GvHD induction and T cell function was assessed in vitro and/or in vivo. Results. By knocking out select HLA class I and class II proteins, we were able to avoid host CD4- and CD8-T cell-mediated recognition. Edited T cells were protected from host NK cells, both in vitro and in an in vivo model engrafted with functional human NK cells. TRAC edited donor T cells did not induce GvHD in an immune compromised mouse model over the 90-day evaluation period. Using our proprietary T cell engineering process, we successfully generated allogeneic T cells with sequential KOs and insertion of a tumor-specific TCR or CAR with high yield. Importantly, these allogeneic T cells had comparable functional activity to their autologous T cell counterparts in in vitro assays (tumor cell killing and cytokine release) as well as in vivo tumor models. With a relatively small bank of donors, we can provide an "off-the-shelf" CAR or TCR-T cell solution for a large proportion of the population. Conclusions. We have successfully developed a differentiated "off-the-shelf" approach, which is expected to be safe and cost-effective. It is designed to provide long-term persistence without the need for an immune suppressive regimen. This promising strategy is being applied to our T cell immuno-oncology and autoimmune research candidates. Disclosures Zhang: Intellia Therapeutics: Current Employment. Goel: Intellia Therapeutics: Current Employment. Prodeus: Intellia Therapeutics: Current Employment. Jetley: Intellia Therapeutics: Current Employment. Tan: Intellia Therapeutics: Current Employment. Averill: Intellia Therapeutics: Current Employment. Ranade: Intellia Therapeutics: Current Employment. Balwani: Intellia Therapeutics: Current Employment. Dutta: Intellia Therapeutics: Current Employment. Sharma: Intellia Therapeutics: Current Employment. Venkatesan: Intellia Therapeutics: Current Employment. Liu: Intellia Therapeutics: Current Employment. Roy: Intellia Therapeutics: Current Employment. O′Connell: Intellia Therapeutics: Current Employment. Arredouani: Intellia Therapeutics: Current Employment. Keenan: Intellia Therapeutics: Current Employment. Lescarbeau: Intellia Therapeutics: Current Employment. Schultes: Intellia Therapeutics: Current Employment.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1929-1929
Author(s):  
Esa Jantunen ◽  
Ville Varmavuo ◽  
Taru Kuittinen ◽  
Tapio Nousiainen ◽  
Pentti Mäntymaa

Abstract Abstract 1929 A combination of chemotherapy plus G-CSF (chemomobilization) is commonly used to mobilize CD34+ cells to circulation. Mobilization of CD34+ cells is poor or suboptimal in 20–30 % of patients. Plerixafor, a CXCR4 antagonist, increases the mobilization of CD34+ cells and may also have effect on graft composition subsequently collected. There are no data on lymphocyte subsets in the grafts collected after chemomobilization plus pre-emptively given plerixafor. We have analyzed lymphocyte subsets (CD3, CD4, CD8, NK cells, CD19) in grafts collected on the next morning after plerixafor injection in 13 chemomobilized patients with non-Hodgkin lymphoma. As controls we had the first collections from 13 NHL patients mobilized with chemotherapy plus G-CSF and with yield of 2–6 × 106/kg CD34+ cells with 1–2 aphaereses. The median CD34+ content of the analyzed grafts was 1.45 × 106/kg in the plerixafor group compared to 1.8 × 106/kg in the controls (p=n.s.). The number of T-cell subsets and NK cells were significantly higher in plerixafor mobilized grafts (Table 1). CD19+ B cells were infrequent in both groups.Table 1.Lymphocyte subsets of the grafts.Stem cell collection with plerixafor, median (range)Stem cell collection without plerixafor, median (range)Significance pGraft volume (ml)100 (43–190)80 (45–140)0.280Graft sample preservation time (days)299 (31–450)291 (103–397)0.898CD34+ cell content (x 106/ kg) after 7-AAD1.45 (0.40–4.40)1.80 (0.31–4.74)0.858CD3+ cell content (x 106/kg)75.3 (14.6–327.3)21.3 (9.1–159.4)0.004CD4+ cell content (x 106/kg)32.7 (10.6–132.8)12.4 (6.9–51.5)0.002CD8+ cell content (x 106/kg)33.4 (4.2–200.5)8.8 (2.2–125.0)0.006CD19+ cell content (x 106/kg)0 (0–0)0 (0–0)NANK cell content (x 106/kg)5.1 (0.2–30.40)1.5 (0.3–8.0)0.045CD4+/CD8+ cell ratio0.98 (0.34–3.04)1.41 (0.28–5.06)0.2287-AAD, 7-Aminoactinomycin D; NK, natural killer. All except one patient has received high-dose therapy with blood stem cell support. The median CD34+ cell dose was 3.1 × 106/kg in plerixafor treated group and 3.3 × 106/kg in the control group, respectively. Time to neutrophil engraftment was comparable between the groups. There were two patients in the plerixafor group with late platelet engraftment (1 and 6 months). Addition of plerixafor to chemomobilization in poor mobilizers results in increased content of T lymphocytes and NK cells in the graft but do not appear to mobilize B lymphocytes. Whether higher T cell and NK cell content are associated with more rapid immune reconstitution and survival should be evaluated in larger patient series with longer follow-up. Disclosures: Jantunen: Genzyme: Honoraria. Kuittinen:Roche: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document