Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors

Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors

ACCEPTED ARTICLE PREVIEW Accepted Article Preview: Published ahead of advance online publication TRANSLATIONAL IMPLICATIONS FOR OFF-THE-SHELF IMMUNE ...

1MB Sizes 2 Downloads 21 Views

ACCEPTED ARTICLE PREVIEW

Accepted Article Preview: Published ahead of advance online publication TRANSLATIONAL IMPLICATIONS FOR OFF-THE-SHELF IMMUNE CELLS EXPRESSING CHIMERIC ANTIGEN RECEPTORS

Hiroki Torikai, and Laurence J.N. Cooper

us

cr ip

t

Cite this article as: Hiroki Torikai, and Laurence J.N. Cooper, TRANSLATIONAL IMPLICATIONS FOR OFF-THE-SHELF IMMUNE CELLS EXPRESSING CHIMERIC ANTIGEN RECEPTORS, Molecular Therapy accepted article preview online 16 May 2016; doi:10.1038/mt.2016.106

Ac

ce

pt

ed

m

an

This is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication. NPG is providing this early version of the manuscript as a service to our customers. The manuscript will undergo copyediting, typesetting and a proof review before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers apply.

Received 03 March 2016 ; accepted 28 April 2016 ; Accepted article preview online 16 May 2016

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Article title TRANSLATIONAL IMPLICATIONS FOR OFF-THE-SHELF IMMUNE CELLS EXPRESSING CHIMERIC ANTIGEN RECEPTORS Short title: Off-the-shelf immune cells expressing CAR Authors: Hiroki Torikai1, and Laurence J.N. Cooper1,2 Author affiliation: Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX

2

Ziopharm Oncology Inc. Boston, MA

cr ip

t

1

us

*Co-corresponding Authors:

an

Laurence J.N. Cooper, M.D., Ph.D.

m

Division of Pediatrics, The University of Texas MD Anderson Cancer Center

ed

Unit 907, 1515 Holcombe Blvd., Houston, TX 77030 Phone: (713) 563-3208; Fax: (713) 792-9832

ce

Hiroki Torikai, M.D.

pt

E-mail: [email protected] and [email protected]

Ac

Division of Pediatrics, The University of Texas MD Anderson Cancer Center Unit 907, 1515 Holcombe Blvd., Houston, TX 77030 Phone: (713) 792-8195; Fax: (713) 792-9832 E-mail: [email protected]

Word count for text: 4481 words, Word count for abstract: 200 words, Figure count: 2 figures, Table Count: 4 tables, Reference count: 133 references

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

ABSTRACT Chimeric antigen receptor (CAR) endows specificity to T-cells independent of HLA. This enables one immunoreceptor to directly target the same surface antigen on different subsets of tumor cells from multiple HLA-disparate recipients. Most approaches manufacture individualized CAR+T-cells from the recipient or HLA-compatible donor, which are revealing promising clinical results. This is the impetus to broaden the number of patients eligible to benefit from adoptive immunotherapy such as to infuse third-party donor derived CAR+T-cells. This will overcome issues associated with (i) time to manufacture T-cells, (ii) cost to generate one product for one patient, (iii) inability to generate a product

cr ip

t

from lymphopenic patients or patient’s immune-cells fail to complete the manufacturing process, and (iv) heterogeneity of T-cell products produced for or from individual recipients. Establishing a bio-bank

us

of allogeneic genetically modified immune-cells from healthy third-party donors, that are cryopreserved

an

and validated in advance of administration, will facilitate the centralizing manufacturing and widespread

m

distribution of CAR+T-cells to multiple points-of-care in a timely manner. To achieve this, it is

ed

necessary to engineer an effective strategy to avoid deleterious allogeneic immune responses leading to

ce

pt

toxicity and rejection. We review the strategies to establish “off-the-shelf” donor-derived bio-banks for

Ac

human application of CAR+T-cells as a drug.

KEY WORDS Off-the-shelf CAR+T-cell, genome editing, allogeneic immune reaction

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

TEXT Introduction The chimeric antigen receptor (CAR) is an artificial immune-receptor to redirect T-cell specificity to tumor-associated cell-surface molecules independent of HLA. The extracellular antigenrecognition domain of the prototypical CAR uses a single chain variable fragment (scFv) from monoclonal antibody (mAb); however this can be replaced with a receptor-ligand interaction of sufficient affinity, such as modified cytokine (e.g., IL-13) to target a cytokine receptor (e.g., IL-13Rα)1, a cell surface molecule (e.g., CD27) for targeting its ligand (e.g., CD70)2, or a pattern-recognition

cr ip

t

receptor (e.g., Dectin-1) for targeting foreign carbohydrates such as β-glucan on germinating Aspergillus3. These binding motifs are typically fused to an extracellular stalk or varying lengths and

an

us

composition, such as derived from the hinge with or without CH2-CH3 domains from IgG1 and IgG4 or

m

hinge with extracellular domain of CD8α. The use of immunoglobulin regions raises the possibility that a CAR may bind to Fc receptors and unwanted elimination of infused T-cells which may be alleviated

pt

_ENREF_8. Recent evidence also suggests the importance of the stalk to impact the effector

ce

6

ed

by introducing site-directed changes or eliminating CH2 region to reduce potential for such clearance4-

Ac

functioning of CAR+T-cells interrogating the binding motifs within TAAs proximal versus distal to the cell surface7-10. CAR-dependent activation is dependent on one or more intracellular signaling domains expressed in cis or trans. The signaling domain of CARs brought forward to clinical applications employed a single signaling molecule containing ITAM motifs in cytoplasmic domain to mimic the TCR/CD3 signal11. These “first generation” CARs typically delivered an incomplete T-cell activation event which prompted embedding additional activation motifs within the CAR endodomain. Inclusion of costimulatory signaling, e.g., through CD2812, CD13713, 14, OX4015, and CD27 domains16, improved function of CAR+T-cells manifested by sustained persistence after adoptive transfer leading to improved therapeutic effect17, 18. In addition to “signal 1” delivered by phosphorylation of ITAMs and “signal 2” mediated by co-stimulatory molecules, T-cells typically require a third signal to achieve and perhaps

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

sustain full activation. This third signal is mediated through the common γ-chain cytokine receptor and thus coordinated delivery or co-expression of certain cytokines can enhance CAR+T-cell functions, which may be especially useful for the application of genetically modified T-cells targeting solid tumors19-21. Human applications of CAR+T-cells have shown promise in several early phase clinical trials, such as infusing targeting CD19 on B-cell leukemias22-30 and lymphomas31-33 and targeting GD2 on neuroblastoma34_ENREF_32. Recently, several reviews have highlighted and summarized these clinical successes35-39. With these early successes, CAR+T-cell therapy is redeploying from academia to

cr ip

t

industry40. Broadening the clinical experience of CAR+T-cell therapy is thus emerging as an important

us

hurdle, especially for those tumor targets already associated with an anti-tumor effect. This underscores

an

the need to expand the list of qualified TAAs that can be safely targeted by T cells though one or more

m

engineered immunoreceptors41. There are a variety of approaches to overcome this, such as to preidentify TAAs expressed solely on tumor cells and targeting aberrant cells based on combinatorial

ed

recognition of more than one TAA,42, 43 or targeting intracellular proteins44, 45.

ce

pt

Another limitation to the broad implementation of CAR+T-cell therapy resides with the

Ac

production process to generate clinical-grade biologics. Currently, T-cells are genetically modified and propagated infusing either patient-derived T-cells often after lympho-depleting chemotherapy or donorderived T-cells administered in the context of hematopoietic stem-cell transplantation (HSCT). These Tcells are produced for a given recipient on a case-by-case basis. This personalized approach to manufacturing reflects the approach pioneered by a subset of academic institutions that operate cell processing facilities in compliance with current good manufacturing practice (GMP) for phase I and II trials on campuses that are in close proximity to clinical facilities to administer CAR+T-cells46. This leads to heterogeneity of manufacturing methods which provides an intellectually fertile and competitive environment to determine facets of production that result in therapeutically-appealing T-cells. However, the physical alignment of multiple geographically diverse GMP facilities with infusion units can lead to

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

duplication of efforts compounding the financial costs associated with a labor-intensive approach to manipulation and testing of T-cells (Figure 1). This in turn raises the cost of therapy and curtails the availability of CAR+T-cells as these products are produced in a limited number of production suites after the recipient has been identified. Moreover, the variability inherent to manufacturing CAR+T-cells from different patients and donors may confound an assessment of mechanisms associated with therapeutic success and complicate the implementation of combination therapies such as with other immune-based modalities. These challenges provide the impetus to develop and implement “off-the-shelf” (OTS) cellbased biologic therapies in which immune cells can be manufactured ahead of need (in advance of

cr ip

t

consent) and infused on demand as required by the recipient, rather than when the biologic is produced.

us

“Off-the-shelf” (OTS) CAR+T-cell therapy

an

The current production and lot release (quality control) of clinical-grade genetically modified T-

m

cells requires time in culture within GMP-compatible manufacturing facilities during which the intended

ed

recipient’s condition can deteriorate. Currently, the “distributed” approach to manufacturing by

ce

pt

academic centers at multiple point-of-care has resulted in a portfolio of biologic products that differ in

Ac

terms of quality and quantity (Table 1) which may impact the results of immunotherapy of CAR+T-cells. OTS immunotherapy may overcome these limitations as it lends itself to centralized manufacture of a well-characterized product. Our present definition of OTS CAR+T-cells is defined as a biologic that is pre-prepared in advance from one or more healthy unrelated donors, validated, and cryopreserved. Their manufacture can be readily undertaken in a centralized manufacturing facility for pre-deployment to treatment facilities and infused as needed rather than when the product is available (Figure 1). One or more biobanks of the OTS T-cell product(s) could then be administered into multiple recipients in multi-center trials powered for efficacy and to establish the maximum tolerated dose. The administration of a wellcharacterized product will then facilitate combination immunotherapies infusing OTS T-cells with other

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

treatments (Table 2). To prepare the cells in advance, we propose the use of “universal” allogeneic donor-derived T-cells. These third party cells might be genetically modified, and as required genetically edited, to safely maintain CAR-mediated effector functions and sustain in vivo persistence by avoiding deleterious immune-mediated recognition by the recipient of allogeneic features on the product (Figure 2).

Strategies to avoid graft-versus-host-disease after infusion of OTS CAR+T-cells In the setting of an HLA-mismatch between donor and recipient, the frequency of T-cells

cr ip

t

specific for disparate HLA is estimated around 1 in 104 47, 48. In clinical trials, the number of administered CAR+T-cells is typically between 108 to 109 which could lead to the delivery 103 to 105 T-

us

cells expressing αβTCR specific for allogeneic antigens and thus likely be sufficient to induce graft-

an

versus-host-disease (GvHD). We describe five strategies to prevent this un-wanted activation through

1. Depletion of alloreactive T-cells

ed

m

endogenous TCR (Table 3).

ce

pt

T-cells displaying αβTCRs play a central role inducing GvHD and therefore several strategies

Ac

have been developed to ex vivo remove alloreactive T-cells such as contaminating the co-infusion of HLA-mismatched hematopoietic stem-cells (HSC) to restore hematopoiesis without GvHD in the context of HSCT. These include the numeric depletion of T-cells that express one or more cell-surface markers consistent with activation (e.g., CD2549-51, CD6952, and CD13753) upon co-culture with antigenpresenting cells (APC) derived from the intended recipient. Activated T-cells can be then be reduced using magnetic beads or immunotoxin conjugated to mAb52,49. Photo-depletion is an alternative approach for reduction of alloreactive T-cells based on the inability of activated T-cells to efflux of a phototoxic dye54. Such methods have already tested in the clinic and significantly decreased the frequency of alloreactive T-cells while preserving viral- and tumor-specific T-cells which may benefit immune reconstitution in an immunocompromised recipient. However, strategies that rely on ex vivo

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

depletion cannot completely eliminate alloreactive T-cells. Moreover, the requirement to co-culture the CAR+T-cells with recipient’s cells reduces the speed and convenience associated with producing this OTS biologic. 2. Allo-anergization of T-cells We demonstrated that anergization of CAR+T-cells can be achieved in tissue culture by combining allo-stimulation with HLA-mismatched APC and concomitant blockade of CD28-mediated co-stimulation55. This resulted in the reduction of recognition of disparate HLA by third-party T-cells mediated by αβTCR while preserving CAR-mediated effector function. The induction of allo-

cr ip

t

anergization requires patient-derived stimulator cells which undermines its suitability for OTS CAR+T-

us

cells.

an

3. T-cells expressing αβTCR with limited or defined specificity

m

One strategy to reduce TCR diversity and thus potential of alloreactivity is to employ T-cells from memory pools as a cellular template for introduction of CAR. Injecting naïve T-cells induced

ed

GvHD in a mouse model, whereas, administering memory T-cells did not56, 57_ENREF_29. This may be

ce

pt

due to a difference in the αβTCR diversity between naïve and memory pools as revealed by Vβ CDR3

Ac

spectratyping or sequencing58, 59. There may also be a functional advantage as mouse memory T-cells could respond to alloantigen, but could not maintain a proliferative response which thus blunted GvHD60. This may have a human application as naïve T-cells can be depleted by recognition of CD45RA while preserving memory T-cells (and HSC)61. The therapeutic potential of adoptive immunotherapy appears to correlate with T-cells expressing a less-differentiated phenotype62 and the sustained numeric expansion of a T-cell subset derived from memory pools to achieve a sizeable biobank may undermine this approach to OTS CAR+T-cell therapy. Enthusiasm for their clinical translation is also undermined by a recent report that failed to show a reduction of acute GvHD using the strategy to deplete naïve population from allogeneic graft 63.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Using the T-cells expressing a defined antigen specificity can curtail the TCR diversity. Adoptive T-cell therapy against a defined peptide/HLA complex should not cause GvHD so long as the restricting αβTCR fail to recognize allogeneic antigens. This is the premise behind clinical trials infusing virus-specific T-cells isolated and expanded from third-party donors to successfully treat opportunistic viral diseases in immunocompromised hosts based on matching at least one HLA allele between donor and recipient that expresses an immunodominant antigen64-67. Administering T-cells expressing αβTCR with defined specificity may be advantageous, as after genetic modification, they will have dual specificity against cell surface TAA (through CAR) and peptide/HLA complex (through

cr ip

t

endogenous TCR). An early-phase clinical trial has demonstrated that this can be accomplished in HLA-

us

matched settings as donor-derived viral-specific CAR+T-cells were infused after allogeneic HSCT27.

an

However, caution may be warranted when delivering antigen-specific T-cells to completely HLA-

m

mismatched recipients as TCR-mediated recognition of target antigens is more promiscuous than

ed

anticipated68. Although GvHD has not been observed after infusing third party HLA-mismatched viral-

ce

substrate for OTS CAR+T-cells.

pt

specific T-cells69, further work is warranted before such antigen-specific T-cells can be used as a cellular

(i) γδT-cells

T-cells

Ac

4. Effector cells other than peripheral blood-derived

Compared to αβTCR, the diversity of γδTCR chain usage is limited which hints that this T-cell population may be less prone to alloreactivity. Although the specificity of some specific γδ T-cells has been identified, the target antigens of most γδTCR are not well characterized and the impact of donorderived γδ T-cells in GvHD pathology is uncertain 70, 71,72, 73. We and others showed that ex vivo activated or expanded human γδ T-cell did not apparently cause xeno-GvHD74-77. Furthermore, the early engraftment of HLA-haploidentical γδ T-cells after infusion of HSC stripped of contaminating αβTCR did not lead to GvHD in humans78. The specificity of one subset of γδ T-cells expressing γ9δ2 TCR

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

recognizes isopentenylpryophosphate (IPP) which can specifically propagated by co-culturing with clinical-grade aminobisphosphonate based upon the inhibition of cholesterol synthesis leading to the accumulation of IPP79. The γ9δ2 T-cell subset can recognize several kinds of tumor cells although to date there has been only a marginal beneficial effect of this subpopulation in clinical trials80. The γ9δ2 T-cell may be combined with CAR to be an option for establishing OTS CAR+T-cell therapy81. However, stimulation(s) with aminobisphosphonate may lead to terminal differentiation which could curtail in vivo persistence. Thus, methodologies to propagate γδ T-cells, including those with polyclonal TCR repertoire, such as using K562-derived artificial antigen presenting cells (aAPC) may be an

cr ip

t

appealing approach to generate OTS CAR+T-cells76. (ii) NKT-cells

an

us

The NKT-cell is a relatively uncommon circulating immune cell that expresses a unique TCR species (Vα24Jα18 in humans). An inverse correlation of recovery of NKT-cells and GvHD has been

m

reported after allogeneic HSCT while preserving a graft-versus-tumor (GVT) response82_ENREF_68.

pt

ed

NKT-cell can also be expanded in vitro by aAPC83 and can be genetically modified to express CAR84.

ce

Thus, the limited TCR usage and emerging technology to obtain large numbers raises the possibility that

(iii) NK-cells

Ac

NKT-cells can be used to generate OTS CAR+ immune cells.

These cells from the innate immune system express a constellation of inhibitory and activation receptors which in aggregate determine the potential for NK-cell mediated cytotoxicity85. An anti-tumor response attributed to NK-cells has been well documented in the context of HLA-mismatched HSCT86-89. To broaden this clinical effect, haploidentical NK-cells have been isolated and infused to treat relapse of acute myelogenous leukemia after lympho-depleting chemotherapy apparently without causing GvHD89, 90

. Despite success in leukemia, currently the evidence that NK-cells can target solid tumor cells in

humans is limited. However, adoptive NK-cell therapy may be advanced through genetic manipulation and/or combining with immune modulators (e.g., lenalidomide and cytokines) and targeting molecules

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

(e.g., therapeutic mAbs participating in antibody-dependent cell-mediated cytotoxicity). Ex vivo numeric expansion of NK-cells can be achieved with recursive additions of γ-irradiated K-562-derived aAPC coexpressing 4-1BBL and membrane bound IL-1591 or IL-2192. Furthermore, the specificity of NK-cells can be directed through enforced expression of a CAR91. These observations lay the foundation for deploying NK-cells as OTS CAR+ immunotherapy. However, a surprising clinical result infusing ex vivo expanded HLA-matched unrelated donor NK-cells revealed an unexpectedly high rate of clinicallysignificant GvHD93. Thus, until additional clinical experience is forthcoming and the mechanism for this adverse event is understood, caution is warranted using allogeneic NK-cells as OTS therapy.

cr ip

t

(iv) Precursor T-cells

T-cells can be generated in vitro by co-culturing CD34+ HSC on Notch-1 ligand-expressing

an

us

mouse stroma cells (OP9/DLL-194 or OP9/DLL-495). This system generates precursor T-cells prior to

m

thymic selection in vivo. MHC-mismatched precursors have been infused into mice with normal thymic function and did not cause GvHD presumably because T-cells expressing TCRs that recognize self-

ed

antigens were eliminated by host thymic selection96. This reveals an approach to administer OTS CAR+

ce

pt

precursor T-cells generated in vitro by partially differentiating HSC with Notch-1 ligand97. As human

Ac

HSC can apparently be safely genetically modified with lentivirus to express CAR98, this approach to OTS immunotherapy may be rendered suitable for human applications. A drawback includes the scale up of ex vivo programming T-cells from HSC to sufficient numbers for biobanking. Furthermore, this strategy might not be suitable for recipients with an immediate need to control of underlying tumor burden due to the time needed in vivo for precursor CAR+T-cells to differentiate into effector T-cells99. (v) Conditional ablation of T-cells Haploidentical T-cells have been genetically modified to express a suicide gene and infused to deliver a GvT-effect after HSCT with the expectation that GvHD can be controlled by eradicating infused T-cells using a pro-drug. The two suicide genes chiefly assessed in clinical trials are thymidine kinase (TK) from herpes simplex virus 1100 and induced caspase 9 (iCasp9)101. An advantage of TK is

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

the ready availability of ganciclovir as a FDA-approved product100,

102, 103

. Disadvantages are the

immunogenicity of viral-derived TK leading to recognition and elimination by the recipient104 and possibly the time course to destroy T-cells after delivery of ganciclovir. An alternative system uses a chemical dimerizer to cross-link iCasp9 leading to cell death. Since iCasp9 is derived from the human gene there is likely reduced potential to elicit immune-mediated recognition and thus deletion. The clinical application of iCasp9 has been published105, 106_ENREF_96 demonstrating resolution of GvHD after infusing haploidentical iCasp9+T-cells were rapidly eliminated by a single infusion of clinicalgrade dimerizer. The clinical appeal of OTS CAR+T-cells that co-express a suicide gene107 will depend

5. Elimination of expression of endogenous

us

TCR

cr ip

t

on the kinetics of elimination and the associated potential for loss of the anti-tumor effect.

an

Artificial nucleases are harnessed ex vivo as genome-editing tools to permanently disrupt the expression of endogenous genes. Zinc finger nucleases (ZFNs) are currently the most clinically mature

m

of the artificial nucleases108. Alternative artificial nucleases, such as transcription activator-like effector

ed

nuclease (TALEN)109, 110 and CRISPR/Cas9 (clustered regularly interspaced short palindromic

ce

pt

repeat/CRISPR-associated proteins)111, 112 are recent technologies that may have translational appeal. We

Ac

applied ZFNs to eliminate expression of endogenous αβTCR from CAR+T-cells thereby completely eliminating the possibility to induce GvHD113. Transient expression of a pair of ZFNs from electrotransferred in vitro-transcribed mRNA resulted in disruption of TCR expression on T-cells genetically modified using the Sleeping Beauty (SB) system to express a CD19-specific CAR. The TCRnegCAR+Tcells were readily enriched by magnetic depletion of the remaining CD3+ population using an approach amenable to clinical translation. Significantly, these genetically edited and modified T-cells could be numerically expanded on CD19+ aAPC. One concern regarding the human application of T-cells genetically edited with artificial nucleases is the occurrence of off-target introduction of double-stranded breaks and the potential for translocation. These must be avoided to exclude genotoxicity and potentially detrimental effects arising after administration of TCRnegCAR+ OTS T-cells. TALEN or CRISPR/Cas9

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

systems may improve off-target-cleavage and also the efficiency of disrupting αβTCR from T-cells114. Using these editing technologies, we have established that the manufacturing process can generate at least 3 x 109 TCRnegCAR+ OTS T-cells from one frozen cord blood unit, which typically contains about 8 x 107 CD3+ T-cells (unpublished data). We estimate that upon completing this propagation and given an infusion dose of 3.0 x 108 per patient29, one can prepare CAR+ OTS T-cells for up to 10 patients within 4 weeks from a single cord blood unit although recurrent stimulation may lead to T-cells with more differentiated phenotype and shortened telomeres that thus may not be suitable for effective adoptive CAR T-cell therapy62. Another round of ex vivo numeric expansion of TCRnegCAR+ OTS T-

cr ip

t

cells will typically increase the cell number 10- fold, which will result in sufficient product for injection

an

leukapheresis product harvested from a healthy donor.

us

into up to 100 patients. This number might be increased upon harvesting T-cells from a steady-state

ed

m

Strategies to sustain persistence of OTS CAR+T-cells in vivo Monoclonal antibodies have found favor as an OTS therapy as they can be readily deployed and simply

ce

pt

infused. Yet, CAR+T-cells possess inherent advantages over mAbs due to their home to sites of

Ac

malignancy, migrate through tissues despite elevated interstitial pressures, increase in numbers, and serially kill target tumor cells. Furthermore, infused CAR+T-cells may survive over the long-term and play an unassisted role in immuno-monitoring to prevent recurrence. In the autologous setting, genetically modified T-cells can survive in human for over 10 years115. However, infusing allogeneic CAR+T-cells from just one unrelated and HLA-disparate donor could activate the recipients’ immune systems and be rejected before an anti-tumor effect is fully realized and prevent long-term immunoprotection. Thus, strategies to avoid clinically-deleterious immune-remediated rejection of OTS CAR+Tcells are needed. In some instances, when the recipient is heavily immunosuppressed, the impact of immune-mediated rejection may not compromise the therapeutic effect such as revealed when 3rd party OTS viral-specific T-cells, matched at only one HLA, are successfully infused to control EBV+

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

lymphoproliferative disease after allogeneic HSCT67. If an HLA-restricted rejection response does arise, then OTS T-cells might be generated as biobanks from more than one donor that are HLA distinct from each other and thus be accessed for the recipient to receive more than one infusion. 1. Suppression of immune system in recipients The depletion of resident immune cells before adoptive transfer of T-cells is generally accepted to enhance the therapeutic effect116, 117. In the context of infusion of allogeneic T-cells this immunosuppression may limit the emergence and/or potency of host-mediated immune response against HLAdisparate OTS CAR+T-cells. One approach to inducing immuno-suppression is based on using

cr ip

t

chemotherapy (e.g., cyclophosphamide and fludarabine) to lympho-deplete the recipient. For example, the elimination of CD52 from CAR T-cells by TALEN may avoid immune suppression by CD52-

us

specific mAb118. This strategy may be used to sidestep clearance of infused OTS CAR+T-cells by an

an

immunosuppressive agent while helping to avoid rejection by the recipient’s immune system. The

m

degree of lymphodepletion achieved with more intensive regimens appears to correlate with improved

ed

persistence of infused T-cells and NK-cells119, 120. What is not known is whether the depth of such

ce

pt

lymphodepletion will lead to sufficient suppression of the immune system to favorably prevent immune-

Ac

mediated rejection of HLA-disparate immune cells. The length of time for the recipient to remain immunosuppressed to achieve a desired CAR-mediated anti-tumor response remains to be determined and during this period the patient is at risk from opportunistic infection. . The human application of OTS T-cells may be useful when combined with allogeneic HSCT and the resultant immunocompromised clinical state. Indeed, an apparently successful infusion of OTS CAR+T-cell therapy using this strategy was recently announced in one recipient, though follow-up is short121. 2. Modulation of immune system in recipients Small molecules, such as sphingosine-1 receptor agonist, FTY720, may be used as modulators of immune system function rather causing immune-mediated suppression. Thus, FTY720 can preclude Tcells from infiltrating into a transplanted allograft which would lead to rejection122, as well as preventing

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

rejection of administered allogeneic CAR+T-cells123, and preventing GvHD in the context of allogeneic HSCT in mice124. The mechanism of suppressing GvHD includes not only inhibition of egress, but apparently also induction of apoptosis of alloreactive T-cells in lymph nodes (LNs)124. It is likely that continuous exposure to FTY720 will be needed to maintain a beneficial effect125, which may adversely affect immune cells other than alloreactive T-cells. Furthermore, since normal B-cells reside within LNs, this might induce the unwanted apoptosis of CD19-specific OTS CAR+T-cells residing in such lymphoid spaces. Thus, FTY720 may prevent adverse events arising from donor-derived OTS CAR+T-cells recognizing recipient alloantigens as well as limiting host-derived T-cells deleteriously recognizing

the potential benefit on survival of infused OTS T-cells.

us

3. Enforced expression of immune checkpoint molecules

cr ip

t

disparate HLA on infused OTS cells. However, safeguarding the specificity of the CAR may preclude

an

Blockade of immune checkpoints can be used to activate resident tumor-specific T-cells to

m

achieve clearance of tumors. Enforced expression of PD-L1 and CTLA4-Ig in human embryonic stem

ed

cells has successfully suppressed rejection of derived allogeneic cells in mice reconstituted with

ce

pt

elements of the human immune system126. Furthermore, allogeneic pancreatic cells differentiated from

Ac

human embryonic stem cells can engraft in humanized mice127. However, the expression of PD-L1 and CTLA4-Ig on CAR+T-cells will presumably result in suppression of effector functions and thus this approach may not be suitable for OTS T-cell therapy. 4. HLA homozygous donors One approach to minimize the rejection of introduced allogeneic cells is to match HLA type between one or more recipients and one or more donors. However, the probability to find a suitable HLA-matched donor for an un-related patient is low as even millions registered with the National Marrow Donor Program cannot provide coverage for the entire U.S. population128. One approach to decrease the number of donors is to use individuals that are homozygous at one or more HLA alleles. For instance, 50 unique donors with HLA homozygous at HLA-A/B/DRB1 are calculated to provide

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

73% of the Japanese population with HLA-matched 3rd party cells and these donors can be found by screening 37,000 Japanese129. In the United Kingdom, 50 donors homozygous for the most prevalent HLA alleles can provide approximately 80% of the population with HLA-matched allogeneic cells130. Therefore, the establishment of a biobank that is homozygous at HLA loci is anticipated to generate OTS CAR+T-cells that are HLA-matched with multiple recipients. In addition to the matching of HLAA/-B/-DRB1, we may need to explore the importance of matching other HLA loci (HLA-C/-DPB1) in OTS T-cell therapeutic settings, as these molecules have been identified as the important alleles related to allogeneic immune responses in allogeneic HSCT setting. Furthermore, the potential clinical

cr ip

t

importance of minor histocompatibility antigen(s) in the mis-matched setting needs to be addressed. One potential source for obtaining HLA homozygous donor derived T-cells would be frozen cord blood units

us

in the public cord blood bank, as HLA information is usually available in those cord blood units and

an

these cells are ready to use.

m

5. Genetic editing of HLA

ed

We have developed an approach to bio-engineering T-cells to limit the number of HLA-

ce

pt

homozygous donors needed to match to an HLA-diverse population. Indeed, designer ZFNs have been

Ac

used to eliminate expression of HLA-A from CAR+T-cells and also TCRnegCAR+T-cells131. One area of possible concern is deleterious NK-cell mediated attack upon recognition of infused HLAnegT-cells. However, this may have limited clinical impact as we have demonstrated that the enforced expression of non-classical non-polymorphous HLA molecules, such as HLA-E or HLA-G, can suppress lysis of genetically edited T-cells by NK-cells circulating in peripheral blood131. Another strategy to avoid NKcell attack may to enforce expression of Siglec-7 and -9 ligands on HLAnull cells132.

Conclusion Autologous CAR+T-cells have demonstrated dramatic anti-tumor responses especially in patients with B-cell tumors. The human applications of OTS CAR+T-cells currently serves as a type of cellular

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

template for testing CAR species that can recognize hematologic tumors. However, these clinical data also reveal challenges (Table 4) to widen their human application within adoptive immunotherapy. For example, we recognize that the initial embodiments of genetically reprogrammed T-cells sourced from a limited number of third party HLA-mismatched donors may be rejected. This will likely be lessened after immune suppression and/or elimination of one or more HLA through genetic editing133. Even so, the presence of minor histocompatibility antigens may still engender an unwanted response in the recipient leading to immune-mediated clearance. The impact of this rejection vector might be blunted by sequentially infusing products derived from more than one donor to achieve a therapeutic response. Thus,

cr ip

t

in the absence of suitable animal models, the anti-tumor activity associated with successive advances in OTS therapies will need to be revealed in iterative clinical trials.

us

In aggregate, targeting of T-cells to antigens independent of HLA provides optimism that

an

CAR+T-cells can be rendered as a “drug”. At this time, OTS approaches belong within a constellation of

m

other immunotherapies such as the adoptive transfer of autologous T cells. For example, during the time

ed

that patient-derived CAR+T-cells are prepared the recipient could receive an OTS cellular therapeutic.

ce

pt

The clinical evaluation of OTS cells for cancer can be contemplated as a modality to provide disease

Ac

control (transient remission) or long-term remission. This distinction will help calibrate the emphasis on bio-engineering methodologies to improve in vivo persistence. The former, while still clinically meaningful, implies that survival of the infused product may be compromised. Whereas, striving for long term remission implies that the OTS product will not be subject to immune-mediated rejection and is HLA compatible with recipient. The decision to infuse autologous or allogeneic CAR+T-cells as monotherapies or in combinations will evolve as the technologies associated with each cellular product is advanced to meet the dual needs of safely and completely eliminating tumor. Overall, the benefits of immediacy, logistics and trial design governing OTS immunotherapies, such as with CAR+T-cells, justify the development of a path to their human applications.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

ACKNOWLEDGEMENT We thank Dr. Judy Moyes for assistance with editing.

AUTHORSHIP Author contribution: H.T. and L.J.N.C. wrote the paper. Conflict of Interest: Some of the technology described in this presentation was advanced through research conducted at the MD Anderson Cancer Center under the direction of Laurence J.N. Cooper. In January 2015, the technology was licensed for commercial application to ZIOPHARM Oncology, Inc.,

cr ip

t

and Intrexon Corporation in exchange for equity interests in each of these companies for which both authors are entitled to receive a portion. On May 7, 2015, Dr. Cooper was appointed as the Chief

Ac

ce

pt

ed

m

an

us

Executive Officer at ZIOPHARM. Dr. Cooper is now a Visiting Scientist at MD Anderson.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

REFERENCES 1.

Brown, CE, Starr, R, Aguilar, B, Shami, AF, Martinez, C, D'Apuzzo, M, et al. (2012). Stem-like tumor-initiating cells isolated from IL13Ralpha2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T Cells. Clin Cancer Res 18: 2199-2209.

2.

Shaffer, DR, Savoldo, B, Yi, Z, Chow, KK, Kakarla, S, Spencer, DM, et al. (2011). T cells redirected against CD70 for the immunotherapy of CD70-positive malignancies. Blood 117: 4304-4314.

3.

Kumaresan, PR, Manuri, PR, Albert, ND, Maiti, S, Singh, H, Mi, T, et al. (2014).

cr ip

t

Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection. Proceedings of the National Academy of Sciences 111: 10660-10665.

Hudecek, M, Sommermeyer, D, Kosasih, PL, Silva-Benedict, A, Liu, L, Rader, C, et al. (2015).

us

4.

an

The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in

m

vivo antitumor activity. Cancer Immunol Res 3: 125-135. Hombach, A, Hombach, AA, and Abken, H (2010). Adoptive immunotherapy with genetically

ed

5.

ce

pt

engineered T cells: modification of the IgG1 Fc 'spacer' domain in the extracellular moiety of

Ac

chimeric antigen receptors avoids 'off-target' activation and unintended initiation of an innate immune response. Gene Ther 17: 1206-1213. 6.

Jonnalagadda, M, Mardiros, A, Urak, R, Wang, X, Hoffman, LJ, Bernanke, A, et al. (2015). Chimeric Antigen Receptors With Mutated IgG4 Fc Spacer Avoid Fc Receptor Binding and Improve T Cell Persistence and Antitumor Efficacy. Mol Ther 23: 757-768.

7.

Hudecek, M, Lupo-Stanghellini, MT, Kosasih, PL, Sommermeyer, D, Jensen, MC, Rader, C, et al. (2013). Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin Cancer Res 19: 3153-3164.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

8.

Jonnalagadda, M, Mardiros, A, Urak, R, Wang, X, Hoffman, LJ, Bernanke, A, et al. (2014). Chimeric Antigen Receptors with Mutated IgG4 Fc Spacer Avoid Fc Receptor Binding and Improve T cell Persistence and Anti-Tumor Efficacy. Mol Ther.

9.

James, SE, Greenberg, PD, Jensen, MC, Lin, Y, Wang, J, Till, BG, et al. (2008). Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol 180: 7028-7038.

10.

Almasbak, H, Walseng, E, Kristian, A, Myhre, MR, Suso, EM, Munthe, LA, et al. (2015). Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse

cr ip

11.

t

model. Gene Ther.

Eshhar, Z, Waks, T, Gross, G, and Schindler, DG (1993). Specific activation and targeting of

us

cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains

an

and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci

Maher, J, Brentjens, RJ, Gunset, G, Riviere, I, and Sadelain, M (2002). Human T-lymphocyte

ed

12.

m

U S A 90: 720-724.

ce

pt

cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat

13.

Ac

Biotechnol 20: 70-75.

Imai, C, Mihara, K, Andreansky, M, Nicholson, IC, Pui, CH, Geiger, TL, et al. (2004). Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18: 676-684.

14.

Milone, MC, Fish, JD, Carpenito, C, Carroll, RG, Binder, GK, Teachey, D, et al. (2009). Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 17: 1453-1464.

15.

Pule, MA, Straathof, KC, Dotti, G, Heslop, HE, Rooney, CM, and Brenner, MK (2005). A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther 12: 933-941.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

16.

Song, DG, Ye, Q, Poussin, M, Harms, GM, Figini, M, and Powell, DJ, Jr. (2012). CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 119: 696-706.

17.

Savoldo, B, Ramos, CA, Liu, E, Mims, MP, Keating, MJ, Carrum, G, et al. (2011). CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 121: 1822-1826.

18.

Carpenito, C, Milone, MC, Hassan, R, Simonet, JC, Lakhal, M, Suhoski, MM, et al. (2009). Control of large, established tumor xenografts with genetically retargeted human T cells

19.

cr ip

t

containing CD28 and CD137 domains. Proc Natl Acad Sci U S A 106: 3360-3365. Hoyos, V, Savoldo, B, Quintarelli, C, Mahendravada, A, Zhang, M, Vera, J, et al. (2010).

us

Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance

Pegram, HJ, Lee, JC, Hayman, EG, Imperato, GH, Tedder, TF, Sadelain, M, et al. (2012).

m

20.

an

their anti-lymphoma/leukemia effects and safety. Leukemia 24: 1160-1170.

ed

Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for

Chinnasamy, D, Yu, Z, Kerkar, SP, Zhang, L, Morgan, RA, Restifo, NP, et al. (2012). Local

Ac

21.

ce

pt

prior conditioning. Blood 119: 4133-4141.

delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin Cancer Res 18: 1672-1683. 22.

Grupp, SA, Kalos, M, Barrett, D, Aplenc, R, Porter, DL, Rheingold, SR, et al. (2013). Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509-1518.

23.

Porter, DL, Levine, BL, Kalos, M, Bagg, A, and June, CH (2011). Chimeric antigen receptormodified T cells in chronic lymphoid leukemia. N Engl J Med 365: 725-733.

24.

Kalos, M, Levine, BL, Porter, DL, Katz, S, Grupp, SA, Bagg, A, et al. (2011). T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3: 95ra73.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

25.

Brentjens, RJ, Davila, ML, Riviere, I, Park, J, Wang, X, Cowell, LG, et al. (2013). CD19targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5: 177ra138.

26.

Brentjens, RJ, Riviere, I, Park, JH, Davila, ML, Wang, X, Stefanski, J, et al. (2011). Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118: 4817-4828.

27.

Cruz, CR, Micklethwaite, KP, Savoldo, B, Ramos, CA, Lam, S, Ku, S, et al. (2013). Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after

28.

cr ip

t

allogeneic stem cell transplant: a phase 1 study. Blood 122: 2965-2973.

Lee, DW, Kochenderfer, JN, Stetler-Stevenson, M, Cui, YK, Delbrook, C, Feldman, SA, et al.

us

(2015). T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in

Maude, SL, Frey, N, Shaw, PA, Aplenc, R, Barrett, DM, Bunin, NJ, et al. (2014). Chimeric

m

29.

an

children and young adults: a phase 1 dose-escalation trial. The Lancet 385: 517-528.

pt

Davila, ML, Riviere, I, Wang, X, Bartido, S, Park, J, Curran, K, et al. (2014). Efficacy and

ce

30.

ed

antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371: 1507-1517.

Ac

toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 6: 224ra225. 31.

Savoldo, B, Ramos, CA, Liu, E, Mims, MP, Keating, MJ, Carrum, G, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 121: 1822-1826.

32.

Kochenderfer, JN, Dudley, ME, Kassim, SH, Somerville, RP, Carpenter, RO, Stetler-Stevenson, M, et al. (2015). Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 33: 540-549.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

33.

Kochenderfer, JN, Yu, Z, Frasheri, D, Restifo, NP, and Rosenberg, SA (2010). Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 116: 3875-3886.

34.

Louis, CU, Savoldo, B, Dotti, G, Pule, M, Yvon, E, Myers, GD, et al. (2011). Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118: 6050-6056.

35.

Batlevi, CL, Matsuki, E, Brentjens, RJ, and Younes, A (2016). Novel immunotherapies in lymphoid malignancies. Nat Rev Clin Oncol 13: 25-40. Sadelain, M (2015). CAR therapy: the CD19 paradigm. J Clin Invest 125: 3392-3400.

37.

Maus, MV, and June, CH (2016). Making Better Chimeric Antigen Receptors for Adoptive T-

Jackson, HJ, Rafiq, S, and Brentjens, RJ (2016). Driving CAR T-cells forward. Nat Rev Clin

an

38.

m

Oncol.

van der Stegen, SJ, Hamieh, M, and Sadelain, M (2015). The pharmacology of second-

ed

39.

us

cell Therapy. Clin Cancer Res 22: 1875-1884.

cr ip

t

36.

June, CH, Riddell, SR, and Schumacher, TN (2015). Adoptive cellular therapy: A race to the

Ac

40.

ce

pt

generation chimeric antigen receptors. Nat Rev Drug Discov 14: 499-509.

finish line. Science Translational Medicine 7: 280ps287. 41.

Torikai, H, Moyes, J, and Cooper, LN (2014). Engineering T Cells to Target Tumor Cells. In: Cai, W (ed). Engineering in Translational Medicine. Springer London. pp 71-101.

42.

Kloss, CC, Condomines, M, Cartellieri, M, Bachmann, M, and Sadelain, M (2012). Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol 31: 71-75.

43.

Fedorov, VD, Themeli, M, and Sadelain, M (2013). PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med 5: 215ra172.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

44.

Inaguma, Y, Akahori, Y, Murayama, Y, Shiraishi, K, Tsuzuki-Iba, S, Endoh, A, et al. (2014). Construction and molecular characterization of a T-cell receptor-like antibody and CAR-T cells specific for minor histocompatibility antigen HA-1H. Gene Ther 21: 575-584.

45.

Chames, P, Willemsen, RA, Rojas, G, Dieckmann, D, Rem, L, Schuler, G, et al. (2002). TCRLike Human Antibodies Expressed on Human CTLs Mediate Antibody Affinity-Dependent Cytolytic Activity. The Journal of Immunology 169: 1110-1118.

46.

Cooper, LJN (2015). Moving from tinkering in the garage to assembly line production: the manufacture of genetically modified T cells expressing chimeric antigen receptors (CARs)

cr ip

47.

t

comes on line. Cancer Gene Therapy.

Heeger, PS (2003). T-cell allorecognition and transplant rejection: a summary and update. Am J

Felix, NJ, and Allen, PM (2007). Specificity of T-cell alloreactivity. Nat Rev Immunol 7: 942-

an

48.

us

Transplant 3: 525-533.

Amrolia, PJ, Muccioli-Casadei, G, Huls, H, Adams, S, Durett, A, Gee, A, et al. (2006). Adoptive

ed

49.

m

953.

ce

pt

immunotherapy with allodepleted donor T-cells improves immune reconstitution after

50.

Ac

haploidentical stem cell transplantation. Blood 108: 1797-1808. Amrolia, PJ, Muccioli-Casadei, G, Yvon, E, Huls, H, Sili, U, Wieder, ED, et al. (2003). Selective depletion of donor alloreactive T cells without loss of antiviral or antileukemic responses. Blood 102: 2292-2299. 51.

Solomon, SR, Mielke, S, Savani, BN, Montero, A, Wisch, L, Childs, R, et al. (2005). Selective depletion of alloreactive donor lymphocytes: a novel method to reduce the severity of graftversus-host disease in older patients undergoing matched sibling donor stem cell transplantation. Blood 106: 1123-1129.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

52.

Hartwig, UF, Nonn, M, Khan, S, Meyer, RG, Huber, C, and Herr, W (2006). Depletion of alloreactive T cells via CD69: implications on antiviral, antileukemic and immunoregulatory T lymphocytes. Bone Marrow Transplant 37: 297-305.

53.

Wehler, TC, Nonn, M, Brandt, B, Britten, CM, Grone, M, Todorova, M, et al. (2007). Targeting the activation-induced antigen CD137 can selectively deplete alloreactive T cells from antileukemic and antitumor donor T-cell lines. Blood 109: 365-373.

54.

Mielke, S, Nunes, R, Rezvani, K, Fellowes, VS, Venne, A, Solomon, SR, et al. (2008). A clinical-scale selective allodepletion approach for the treatment of HLA-mismatched and

cr ip

t

matched donor-recipient pairs using expanded T lymphocytes as antigen-presenting cells and a TH9402-based photodepletion technique. Blood 111: 4392-4402.

Davies, JK, Singh, H, Huls, H, Yuk, D, Lee, DA, Kebriaei, P, et al. (2010). Combining CD19

us

55.

an

redirection and alloanergization to generate tumor-specific human T cells for allogeneic cell

m

therapy of B-cell malignancies. Cancer Res 70: 3915-3924. Anderson, BE, McNiff, J, Yan, J, Doyle, H, Mamula, M, Shlomchik, MJ, et al. (2003). Memory

ed

56.

Foster, AE, Marangolo, M, Sartor, MM, Alexander, SI, Hu, M, Bradstock, KF, et al. (2004).

Ac

57.

ce

pt

CD4+ T cells do not induce graft-versus-host disease. J Clin Invest 112: 101-108.

Human CD62L- memory T cells are less responsive to alloantigen stimulation than CD62L+ naive T cells: potential for adoptive immunotherapy and allodepletion. Blood 104: 2403-2409. 58.

Arstila, TP, Casrouge, A, Baron, V, Even, J, Kanellopoulos, J, and Kourilsky, P (1999). A direct estimate of the human alphabeta T cell receptor diversity. Science 286: 958-961.

59.

Robins, HS, Campregher, PV, Srivastava, SK, Wacher, A, Turtle, CJ, Kahsai, O, et al. (2009). Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood 114: 4099-4107.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

60.

Chen, BJ, Deoliveira, D, Cui, X, Le, NT, Son, J, Whitesides, JF, et al. (2007). Inability of memory T cells to induce graft-versus-host disease is a result of an abortive alloresponse. Blood 109: 3115-3123.

61.

Bleakley, M, Heimfeld, S, Jones, LA, Turtle, C, Krause, D, Riddell, SR, et al. (2014). Engineering human peripheral blood stem cell grafts that are depleted of naive T cells and retain functional pathogen-specific memory T cells. Biol Blood Marrow Transplant 20: 705-716.

62.

Gattinoni, L, Klebanoff, CA, and Restifo, NP (2012). Paths to stemness: building the ultimate antitumour T cell. Nat Rev Cancer 12: 671-684.

t

Bleakley, M, Heimfeld, S, Loeb, KR, Jones, LA, Chaney, C, Seropian, S, et al. (2015).

cr ip

63.

Outcomes of acute leukemia patients transplanted with naive T cell–depleted stem cell grafts.

Haque, T, Wilkie, GM, Jones, MM, Higgins, CD, Urquhart, G, Wingate, P, et al. (2007).

an

64.

us

Journal of Clinical Investigation.

m

Allogeneic cytotoxic T-cell therapy for EBV-positive posttransplantation lymphoproliferative

pt

Haque, T, Wilkie, GM, Taylor, C, Amlot, PL, Murad, P, Iley, A, et al. (2002). Treatment of

ce

65.

ed

disease: results of a phase 2 multicenter clinical trial. Blood 110: 1123-1131.

Ac

Epstein-Barr-virus-positive post-transplantation lymphoproliferative disease with partly HLAmatched allogeneic cytotoxic T cells. The Lancet 360: 436-442. 66.

Barker, JN, Doubrovina, E, Sauter, C, Jaroscak, JJ, Perales, MA, Doubrovin, M, et al. (2010). Successful treatment of EBV-associated posttransplantation lymphoma after cord blood transplantation using third-party EBV-specific cytotoxic T lymphocytes. Blood 116: 5045-5049.

67.

Leen, AM, Bollard, CM, Mendizabal, AM, Shpall, EJ, Szabolcs, P, Antin, JH, et al. (2013). Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 121: 5113-5123.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

68.

Amir, AL, D'Orsogna, LJ, Roelen, DL, van Loenen, MM, Hagedoorn, RS, de Boer, R, et al. (2010). Allo-HLA reactivity of virus-specific memory T cells is common. Blood 115: 31463157.

69.

Melenhorst, JJ, Leen, AM, Bollard, CM, Quigley, MF, Price, DA, Rooney, CM, et al. (2010). Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood 116: 4700-4702.

70.

Chien, YH, and Konigshofer, Y (2007). Antigen recognition by gammadelta T cells. Immunol Rev 215: 46-58.

t

Xi, X, Guo, Y, Chen, H, Xu, C, Zhang, H, Hu, H, et al. (2009). Antigen specificity of

cr ip

71.

gammadelta T cells depends primarily on the flanking sequences of CDR3delta. J Biol Chem

Maeda, Y, Reddy, P, Lowler, KP, Liu, C, Bishop, DK, and Ferrara, JL (2005). Critical role of

an

72.

us

284: 27449-27455.

m

host gammadelta T cells in experimental acute graft-versus-host disease. Blood 106: 749-755. Blazar, BR, Taylor, PA, Panoskaltsis-Mortari, A, Barrett, TA, Bluestone, JA, and Vallera, DA

ed

73.

ce

pt

(1996). Lethal murine graft-versus-host disease induced by donor gamma/delta expressing T

Ac

cells with specificity for host nonclassical major histocompatibility complex class Ib antigens. Blood 87: 827-837. 74.

Drobyski, WR, Vodanovic-Jankovic, S, and Klein, J (2000). Adoptively transferred gamma delta T cells indirectly regulate murine graft-versus-host reactivity following donor leukocyte infusion therapy in mice. J Immunol 165: 1634-1640.

75.

Vodanovic-Jankovic, S, and Drobyski, WR (2006). Gammadelta T cells do not require fully functional cytotoxic pathways or the ability to recognize recipient alloantigens to prevent graft rejection. Biol Blood Marrow Transplant 12: 1125-1134.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

76.

Deniger, DC, Maiti, S, Mi, T, Switzer, K, Ramachandran, V, Hurton, LV, et al. (2014). Activating and propagating polyclonal gamma delta T cells with broad specificity for malignancies. Clin Cancer Res.

77.

Deniger, DC, Switzer, K, Mi, T, Maiti, S, Hurton, L, Singh, H, et al. (2013). Bispecific T-cells expressing polyclonal repertoire of endogenous gammadelta T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol Ther 21: 638-647.

78.

Wilhelm, M, Smetak, M, Schaefer-Eckart, K, Kimmel, B, Birkmann, J, Einsele, H, et al. (2014). Successful adoptive transfer and in vivo expansion of haploidentical gammadelta T cells. J

cr ip

Gober, HJ, Kistowska, M, Angman, L, Jeno, P, Mori, L, and De Libero, G (2003). Human T Cell Receptor

Cells Recognize Endogenous Mevalonate Metabolites in Tumor Cells. Journal of

us

79.

t

Transl Med 12: 45.

Fisher, JP, Heuijerjans, J, Yan, M, Gustafsson, K, and Anderson, J (2014). gammadelta T cells

m

80.

an

Experimental Medicine 197: 163-168.

pt

Fisher, JPH, Flutter, B, Wesemann, F, Frosch, J, Rossig, C, Gustafsson, K, et al. (2015).

ce

81.

ed

for cancer immunotherapy: A systematic review of clinical trials. Oncoimmunology 3: e27572.

Ac

Effective combination treatment of GD2-expressing neuroblastoma and Ewing's sarcoma using anti-GD2 ch14.18/CHO antibody with Vγ9Vδ2+ γδT cells. OncoImmunology: 00-00. 82.

Pillai, AB, George, TI, Dutt, S, Teo, P, and Strober, S (2007). Host NKT Cells Can Prevent Graft-versus-Host Disease and Permit Graft Antitumor Activity after Bone Marrow Transplantation. The Journal of Immunology 178: 6242-6251.

83.

Webb, TJ, Bieler, JG, Schneck, JP, and Oelke, M (2009). Ex vivo induction and expansion of natural killer T cells by CD1d1-Ig coated artificial antigen presenting cells. J Immunol Methods 346: 38-44.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

84.

Heczey, A, Liu, D, Tian, G, Courtney, AN, Wei, J, Marinova, E, et al. (2014). Invariant NKT cells with chimeric antigen receptor provide a novel platform for safe and effective cancer immunotherapy. Blood 124: 2824-2833.

85.

Ljunggren, HG, and Malmberg, KJ (2007). Prospects for the use of NK cells in immunotherapy of human cancer. Nat Rev Immunol 7: 329-339.

86.

Ruggeri, L, Mancusi, A, Capanni, M, Urbani, E, Carotti, A, Aloisi, T, et al. (2007). Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value. Blood 110: 433-440.

t

Moretta, L, Locatelli, F, Pende, D, Marcenaro, E, Mingari, MC, and Moretta, A (2011). Killer Ig-

cr ip

87.

stem cell transplantation. Blood 117: 764-771.

Curti, A, Ruggeri, L, D'Addio, A, Bontadini, A, Dan, E, Motta, MR, et al. (2011). Successful

an

88.

us

like receptor-mediated control of natural killer cell alloreactivity in haploidentical hematopoietic

m

transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in

pt

Rubnitz, JE, Inaba, H, Ribeiro, RC, Pounds, S, Rooney, B, Bell, T, et al. (2010). NKAML: a

ce

89.

ed

elderly high risk acute myeloid leukemia patients. Blood 118: 3273-3279.

Ac

pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol 28: 955-959. 90.

Bachanova, V, Cooley, S, Defor, TE, Verneris, MR, Zhang, B, McKenna, DH, et al. (2014). Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood 123: 3855-3863.

91.

Imai, C, Iwamoto, S, and Campana, D (2005). Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 106: 376383.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

92.

Denman, CJ, Senyukov, VV, Somanchi, SS, Phatarpekar, PV, Kopp, LM, Johnson, JL, et al. (2012). Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS One 7: e30264.

93.

Shah, NN, Baird, K, Delbrook, CP, Fleisher, TA, Kohler, ME, Rampertaap, S, et al. (2015). Acute GVHD in patients receiving IL-15/4-1BBL activated NK cells following T-cell-depleted stem cell transplantation. Blood 125: 784-792.

94.

Awong, G, Herer, E, Surh, CD, Dick, JE, La Motte-Mohs, RN, and Zuniga-Pflucker, JC (2009). Characterization in vitro and engraftment potential in vivo of human progenitor T cells generated

cr ip

95.

t

from hematopoietic stem cells. Blood 114: 972-982.

Mohtashami, M, Shah, DK, Nakase, H, Kianizad, K, Petrie, HT, and Zuniga-Pflucker, JC (2010).

us

Direct comparison of Dll1- and Dll4-mediated Notch activation levels shows differential

Zakrzewski, JL, Kochman, AA, Lu, SX, Terwey, TH, Kim, TD, Hubbard, VM, et al. (2006).

m

96.

an

lymphomyeloid lineage commitment outcomes. J Immunol 185: 867-876.

ed

Adoptive transfer of T-cell precursors enhances T-cell reconstitution after allogeneic

Zakrzewski, JL, Suh, D, Markley, JC, Smith, OM, King, C, Goldberg, GL, et al. (2008). Tumor

Ac

97.

ce

pt

hematopoietic stem cell transplantation. Nat Med 12: 1039-1047.

immunotherapy across MHC barriers using allogeneic T-cell precursors. Nat Biotechnol 26: 453461. 98.

De Oliveira, SN, Ryan, C, Giannoni, F, Hardee, CL, Tremcinska, I, Katebian, B, et al. (2013). Modification of hematopoietic stem/progenitor cells with CD19-specific chimeric antigen receptors as a novel approach for cancer immunotherapy. Hum Gene Ther 24: 824-839.

99.

Gschweng, E, De Oliveira, S, and Kohn, DB (2014). Hematopoietic stem cells for cancer immunotherapy. Immunol Rev 257: 237-249.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

100.

Bonini, C, Ferrari, G, Verzeletti, S, Servida, P, Zappone, E, Ruggieri, L, et al. (1997). HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia. Science 276: 1719-1724.

101.

Straathof, KC, Pule, MA, Yotnda, P, Dotti, G, Vanin, EF, Brenner, MK, et al. (2005). An inducible caspase 9 safety switch for T-cell therapy. Blood 105: 4247-4254.

102.

Bonini, C, Bondanza, A, Perna, SK, Kaneko, S, Traversari, C, Ciceri, F, et al. (2007). The suicide gene therapy challenge: how to improve a successful gene therapy approach. Mol Ther 15: 1248-1252.

t

Ciceri, F, Bonini, C, Stanghellini, MT, Bondanza, A, Traversari, C, Salomoni, M, et al. (2009).

cr ip

103.

Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic

us

stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised phase I-II study.

Berger, C, Flowers, ME, Warren, EH, and Riddell, SR (2006). Analysis of transgene-specific

m

104.

an

Lancet Oncol 10: 489-500.

ed

immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified

Di Stasi, A, Tey, SK, Dotti, G, Fujita, Y, Kennedy-Nasser, A, Martinez, C, et al. (2011).

Ac

105.

ce

pt

donor T cells after allogeneic hematopoietic cell transplantation. Blood 107: 2294-2302.

Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 365: 1673-1683. 106.

Zhou, X, Di Stasi, A, Tey, SK, Krance, RA, Martinez, C, Leung, KS, et al. (2014). Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood 123: 3895-3905.

107.

Gargett, T, and Brown, MP (2014). The inducible caspase-9 suicide gene system as a "safety switch" to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front Pharmacol 5: 235.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

108.

Perez, EE, Wang, J, Miller, JC, Jouvenot, Y, Kim, KA, Liu, O, et al. (2008). Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 26: 808-816.

109.

Miller, JC, Tan, S, Qiao, G, Barlow, KA, Wang, J, Xia, DF, et al. (2011). A TALE nuclease architecture for efficient genome editing. Nat Biotechnol 29: 143-148.

110.

Zhang, F, Cong, L, Lodato, S, Kosuri, S, Church, GM, and Arlotta, P (2011). Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat Biotechnol 29: 149-153.

t

Cong, L, Ran, FA, Cox, D, Lin, S, Barretto, R, Habib, N, et al. (2013). Multiplex genome

cr ip

111.

engineering using CRISPR/Cas systems. Science 339: 819-823.

Mali, P, Yang, L, Esvelt, KM, Aach, J, Guell, M, DiCarlo, JE, et al. (2013). RNA-guided human

us

112.

Torikai, H, Reik, A, Liu, PQ, Zhou, Y, Zhang, L, Maiti, S, et al. (2012). A foundation for

m

113.

an

genome engineering via Cas9. Science 339: 823-826.

ed

universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-

Osborn, MJ, Webber, BR, Knipping, F, Lonetree, CL, Tennis, N, DeFeo, AP, et al. (2015).

Ac

114.

ce

pt

antigen-receptor and eliminate expression of endogenous TCR. Blood 119: 5697-5705.

Evaluation of TCR Gene Editing achieved by TALENs, CRISPR/Cas9 and megaTAL nucleases. Mol Ther. 115.

Scholler, J, Brady, TL, Binder-Scholl, G, Hwang, WT, Plesa, G, Hege, KM, et al. (2012). Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med 4: 132ra153.

116.

Gattinoni, L, Powell, DJ, Jr., Rosenberg, SA, and Restifo, NP (2006). Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol 6: 383-393.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

117.

Gattinoni, L, Finkelstein, SE, Klebanoff, CA, Antony, PA, Palmer, DC, Spiess, PJ, et al. (2005). Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 202: 907-912.

118.

Poirot, L, Philip, B, Schiffer-Mannioui, C, Le Clerre, D, Chion-Sotinel, I, Derniame, S, et al. (2015). Multiplex Genome-Edited T-cell Manufacturing Platform for "Off-the-Shelf" Adoptive T-cell Immunotherapies. Cancer Res 75: 3853-3864.

119.

Wrzesinski, C, Paulos, CM, Kaiser, A, Muranski, P, Palmer, DC, Gattinoni, L, et al. (2010). Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred

cr ip

120.

t

tumor-specific T cells. J Immunother 33: 1-7.

Geller, MA, Cooley, S, Judson, PL, Ghebre, R, Carson, LF, Argenta, PA, et al. (2011). A phase

us

II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast

Couzin-Frankel, J (2015). Baby's leukemia recedes after novel cell therapy. Science 350: 731-

m

121.

ed

731.

pt

Martini, S, Peters, H, Bohler, T, and Budde, K (2007). Current perspectives on FTY720. Expert

ce

122.

an

cancer. Cytotherapy 13: 98-107.

123.

Ac

Opin Investig Drugs 16: 505-518. Marcus, A, and Eshhar, Z (2011). Tumor-specific allogeneic cells for cancer therapy. Expert Opin Biol Ther 11: 1551-1554. 124.

Hashimoto, D, Asakura, S, Matsuoka, K, Sakoda, Y, Koyama, M, Aoyama, K, et al. (2007). FTY720 enhances the activation-induced apoptosis of donor T cells and modulates graft-versushost disease. Eur J Immunol 37: 271-281.

125.

Taylor, PA, Kelly, RM, Bade, ND, Smith, MJ, Stefanski, HE, and Blazar, BR (2012). FTY720 markedly increases alloengraftment but does not eliminate host anti-donor T cells that cause graft rejection on its withdrawal. Biol Blood Marrow Transplant 18: 1341-1352.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

126.

Rong, Z, Wang, M, Hu, Z, Stradner, M, Zhu, S, Kong, H, et al. (2014). An effective approach to prevent immune rejection of human ESC-derived allografts. Cell Stem Cell 14: 121-130.

127.

Szot, GL, Yadav, M, Lang, J, Kroon, E, Kerr, J, Kadoya, K, et al. (2014). Tolerance Induction and Reversal of Diabetes in Mice Transplanted with Human Embryonic-Stem-Cell-Derived Pancreatic Endoderm. Cell Stem Cell.

128.

Gragert, L, Eapen, M, Williams, E, Freeman, J, Spellman, S, Baitty, R, et al. (2014). HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. N Engl J Med 371: 339-348.

129.

Okita, K, Matsumura, Y, Sato, Y, Okada, A, Morizane, A, Okamoto, S, et al. (2011). A more

130.

cr ip

t

efficient method to generate integration-free human iPS cells. Nat Methods 8: 409-412. Taylor, CJ, Peacock, S, Chaudhry, AN, Bradley, JA, and Bolton, EM (2012). Generating an

us

iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA

Torikai, H, Reik, A, Soldner, F, Warren, EH, Yuen, C, Zhou, Y, et al. (2013). Toward

m

131.

an

types. Cell Stem Cell 11: 147-152.

pt

Jandus, C, Boligan, KF, Chijioke, O, Liu, H, Dahlhaus, M, Demoulins, T, et al. (2014).

Ac

132.

ce

122: 1341-1349.

ed

eliminating HLA class I expression to generate universal cells from allogeneic donors. Blood

Interactions between Siglec-7/9 receptors and ligands influence NK cell-dependent tumor immunosurveillance. J Clin Invest 124: 1810-1820. 133.

Torikai, H, Mi, T, Gragert, L, Maiers, M, Najjar, A, Ang, S, et al. (2016). Genetic editing of HLA expression in hematopoietic stem cells to broaden their human application. Scientific Reports 6: 21757.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

FIGURE LEGENDS Figure 1. Manufacturing of CAR+T-cells. a. Current manufacturing of CAR+T-cells: CAR+T-cells are generated and infused into recipient in each facility. b. Proposed manufacturing OTS CAR+T-cells: CAR+T-cells are generated in a single facility and distributed to multiple points-of-care.

t

Figure 2. Schematic presentation of potential issues in establishing OTS CAR+T-cells from one or

cr ip

more third party donors.

us

While allogeneic CAR+T-cells can destruct target tumor cells, they may also recognize patient’s somatic

an

cells through endogenous TCR, which results in the deleterious GvHD. We will need to avoid this

m

allogeneic immune reaction induced by infused allogeneic CAR+T-cells. Further consideration will be

ed

needed to preclude recognition of infused CAR+T-cells by recipient’s immune system to sustain

Ac

ce

pt

CAR+T-cells in vivo persistence.

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Table 1: Heterogeneity in the biologic products as derived from different autologous or allogeneic donors

Number of copies of integrated CAR Genomic sites of transgene insertion(s) T-cell immuno-phenotype TCR repertoire

Ac

ce

pt

ed

m

an

us

cr ip

t

Effector function due to the phenotype, single nucleotide polymorphisms (SNPs), etc…

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Table 2: Advantages associated with infusing OTS CAR+T-cells manufactured from one or more 3rd party donor(s) for administration and re-administration into multiple unrelated recipients. Avoids difficulties in generating CAR+T-cells from and for a recipient due to poor quality and/or quantity of T-cells Reduces the cost, time, and resources to manufacture CAR+T-cells that are needed for infusion into a single patient OTS CAR+T-cells can be pre-prepared for infusion as needed versus when available OTS CAR+T-cells can be pre-deployed at multiple points-of-care

cr ip

t

OTS CAR+T-cells have reduced heterogeneity compared with multiple patient-specific

us

CAR+T-cells

an

OTS CAR+T-cells can be produced using centralized manufacturing site(s) OTS CAR+T-cells reduce the barriers to undertaking Phase IIb multi-center trials powered

m

for efficacy

ed

OTS CAR+T-cells can be infused as part of multi-component trials administering this

Ac

ce

pt

biologic at predefined maximally tolerated dose

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Table3: Potential strategies to generate OTS cell products Avoid GvHD Eliminate alloreactive T-cells CD25, CD69, CD137 depletion, photo-depletion Suppress TCR mediated activation B7 specific mAb for blocking CD28 mediated co-stimulation Decrease the TCR repertoire Memory T-cells T-cells expressing TCR with defined specificity Virus or tumor specific T-cells, γ9δ2 T-cell Use cell types that are not expressing alloreactive immunoreceptors

cr ip

t

γδ T-cells, NK-cells, NKT-cells Use precursor T-cells

us

Conditional ablation

an

HSV1-TK, iCasp9 Eliminate of endogenous TCR

m

ZFN, TALEN, CRISPR/Cas9 and other gene editing technologies

ed

Avoid rejection

Suppress immune system in recipients

pt

lymphodepletion (CY, Fludarabine, TBI, anti-CD52Ab)

Ac

FTY720

ce

Modulation of immune system in recipients

Enforced expression of immune checkpoint molecules PD-L1, CTLA4-Ig HLA homozygous donor Gene editing of HLA ZFN, TALEN, CRISPR/Cas9 and other gene editing technologies

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Table 4: Questions to improve the therapeutic potential and acceptance of infused OTS T-cells in human trials for oncology What degree of HLA matching is needed between a donor and multiple recipients to prevent immune-mediated rejection and deleterious clearance leading to loss of anti-tumor response? What lympho-depleting and/or immune-suppressive regimens are needed to prevent immunemediated rejection and deleterious clearance leading to loss of anti-tumor response? What defines the ideal donor and do these criteria differ based on tumor burden, type, and recipient? How will donor-to-donor variation and the potential to impact the anti-tumor response both be measured/optimized?

t

Will scale up and manufacturing of large bio-banks impact the anti-tumor activity and how will

cr ip

this be a prior assessed?

Should investigators and recipients expect (temporary) disease control rather than (sustained)

us

anti-tumor effects?

an

What clinical context is best suited for initial clinical testing given the variabilities of tumor (i)

m

types, (ii) burdens, and (iii) distributions?

What clinical context is best suited for initial clinical testing given the variabilities of each

ed

recipient’s (i) genetic background, (ii) body mass, and (iii) prior therapies?

pt

How to address the targeting of solid tumors?

ce

Should Phase I trials anticipate that recipients receive more the one infusion?

Ac

Are T cells the preferred cellular substrate for an OTS biologic/product?

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Figure 1

© 2016 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Figure 2

© 2016 The American Society of Gene & Cell Therapy. All rights reserved