ART. Science
ART. Science
CRISPR-Cas9 gene editing provides a powerful tool to enhance the natural ability of human T cells to fight
cancer. We report a first-in-human phase I clinical trial to test the safety and feasibility of multiplex
CRISPR-Cas9 editing to engineer T cells in three patients with refractory cancer. Two genes encoding the
endogenous T cell receptor (TCR) chains, TCRα (TRAC) and TCRβ (TRBC) were deleted in T cells to reduce
TCR mispairing and to enhance the expression of a synthetic, cancer-specific TCR transgene (NY-ESO-1).
Removal of a third gene encoding PD-1 (PDCD1), was performed to improve anti-tumor immunity. Adoptive
transfer of engineered T cells into patients resulted in durable engraftment with edits at all three genomic
loci. Though chromosomal translocations were detected, the frequency decreased over time. Modified
T cells persisted for up to 9 months suggesting that immunogenicity is minimal under these conditions and
demonstrating the feasibility of CRISPR gene-editing for cancer immunotherapy.
Gene editing offers the potential to correct DNA mutations, to HIV infection (3, 4). The incorporation of multiple guide
and may offer promise to treat or eliminate countless human sequences in CRISPR–Cas9 permits, in principle, multiplex
genetic diseases. The goal of gene editing is to change the genome engineering at several sites within a mammalian ge-
DNA of cells with single base pair precision. The principle nome (5–9). The ability of CRISPR to facilitate efficient mul-
was first demonstrated in mammalian cells when it was tiplex genome editing has greatly expanded the scope of
shown that expression of a rare cutting endonuclease to cre- possible targeted genetic manipulations, enabling new possi-
ate double-strand DNA breaks resulted in repair by homolo- bilities such as simultaneous deletion or insertion of multiple
gous and non-homologous recombination (1). A variety of DNA sequences in a single round of mutagenesis. The pro-
engineered nucleases were then developed to increase effi- spect of using CRISPR-engineering to treat a host of diseases
ciency and enable potential therapeutic applications, includ- such as inherited blood disorders and blindness is moving
ing zinc finger nucleases, homing endonucleases, closer to reality.
transcription activator-like effector nucleases and CRISPR– Recent advances in CRISPR-Cas9 technology have also
Cas9 (clustered regularly interspaced short palindromic re- permitted efficient DNA modifications in human T cells,
peats associated with Cas9 endonuclease) (2). The first pilot which holds great promise for enhancing the efficacy of can-
human trials using genome editing were conducted in pa- cer therapy. T lymphocytes are specialized immune cells that
tients with HIV/AIDS and targeted the white blood cell pro- are largely at the core of the modern day cancer immunother-
tein CCR5, with the goal of mutating the gene by non- apy revolution. The T cell receptor (TCR) complex is located
homologous recombination and thereby inducing resistance on the surface of T cells, and is central for initiating
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 1
successful anti-tumor responses by recognizing foreign anti- Results
gens/peptides bound to MHC-molecules. One of the most Clinical protocol
promising areas of cancer immunotherapy involves adoptive The phase I human trial (ClinicalTrials.gov NCT03399448)
cell therapy, where the patient’s own T cells are genetically was designed to assess the safety and feasibility of infusing
engineered to express a synthetic (transgenic) TCR that can autologous NY-ESO-1 TCR engineered T cells in patients after
specifically detect and kill tumor cells. Recent studies have CRISPR-Cas9 editing of the TRAC, TRBC and PDCD1 loci.
shown safety and promising efficacy of such adoptive T cell During the manufacturing process, cells were taken out of the
transfer approaches using transgenic TCRs specific for the cancer patient, engineered and then infused back into the in-
immunogenic NY-ESO-1 tumor antigen in patients with mye- dividual. The genetically-engineered T cell product was
loma, melanoma and sarcoma (10–12). One limitation of this termed “NYCE” (NY-ESO-1 transduced CRISPR 3X edited
approach is that the transgenic TCR has been shown to mis- cells) and is referred to as NYCE hereafter. During clinical
pair and/or compete for expression with the alpha and beta development of the protocol, we elected to use a TCR rather
chains of the endogenous TCR (13–15). Mispairing of the ther- than a CAR because the incidence of cytokine release syn-
apeutic TCR alpha and beta chains with endogenous alpha drome is generally less prevalent using TCRs (11). In princi-
and beta chains reduces therapeutic TCR cell surface expres- ple, this allowed a more discriminating assessment of
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 3
PDCD1 was the most specific, as very few off-target edits were declined in frequency until cell harvest. The translocations
identified in over 7000 sites of cleavage, and there were very and the TRBC1:TRBC2 deletion were evident in the three pa-
few off-target reads identified at the TRAC1 and TRAC2 loci tients between 10 days after infusion and 30 to 170 days after
(Fig. 4C). infusion (Fig. 5B). However, the rearrangements declined in
The genomic localization of identified DNA cleavage sites frequency in vivo suggesting that they conferred no evidence
was as expected, given the chromosomal location of the three of a growth advantage over many generations of expansion
targeted genes on chromosomes 2, 7 and 14 (Fig. 4A). The dis- in the patients on this trial (Fig. 3, A and B). At day 30, 150
tribution of the incorporation of the dsODN label around on- and 170 in patients UPN07, UPN35 and UPN39, chromosomal
target sites, based on pileups within a window of 100 base translocations were at the limits of detection or not detected
pairs, is shown in Fig. 4B and fig. S4. While most mutations for all rearrangements except for the 9.3 kB deletion for
were on target, there were off-target mutations identified TRBC1:TRBC2.
(Fig. 4C and fig. S5). For the TRAC sgRNA, there were low
abundance mutations within the transcriptional unit of Single-cell RNA sequencing analysis reveals evolution
CLIC2 (chloride intracellular channel 2); however disruption of CRISPR-Cas9 engineered NYCE cells
of CLIC2 in T cells is not expected to have negative conse- We used single cell RNA sequencing (scRNAseq) to compre-
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 4
antitumor efficacy in preclinical models (19, 21–24). We found engineered T cells (Fig. 7D).
that ~25% of the T cells expressing the NY-ESO-1 TCR in the
infusion product had mutations in the PDCD1 locus (fig. S8). Discussion
It is interesting that the frequency of cells with edits in the Our phase I first-in-human pilot study demonstrates the ini-
PDCD1 locus decreased to ~5% of the cells expressing the tial safety and feasibility of multiplex CRISPR-Cas9 T cell hu-
transgenic TCR at 4 months after infusion. This would be man genome engineering in patients with advanced,
consistent with mouse studies of chronic infection where refractory cancer. In one patient analyzed at depth, a fre-
PD-1 deficient T cells are less able to establish memory (18). quency of 30% of di-genic and tri-genic editing was achieved
Figure 6D shows the distribution of engineered T cells in in the infused cell population, and 20% of the TCR transgenic
patients expressing the NY-ESO-1 TCR transgene over time, T cells in circulation 4 months later had persisting di-genic
as they evolved from the infusion product at baseline and and tri-genic edits. We chose to redirect specificity of the
then again at 4 months in vivo. In the heat map (Fig. 6E), the fT cells with a T cell receptor, rather than a chimeric antigen
most differentially expressed genes in the cells expressing the receptor, in order to avoid the chimeric antigen receptor-as-
NY-ESO-1 transcript at the various time points are shown in sociated potential toxicities such as cytokine release syn-
table S5. It is notable that this patient had increases in ex- drome (31). This provided a lower baseline toxicity profile,
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 5
between PDCD1:TRAC and TRBC1:TRBC2. The frequency of gRNA was prepared from linearized DNA (Aldevron) using
translocations that we observed with tri-genic editing is sim- Bulk T7 Megascript 5X (Ambion) and purified using RNeasy
ilar to that reported for di-genic editing using transcription Maxi Kit (Qiagen).
activator-like effector nuclease (TALEN)-mediated gene edit-
ing in preclinical and clinical studies, where rearrangements Recombinant Cas9 protein
were detected in approximately 4% of cells (39, 40). It is im- Cas9 recombinant protein derived from Streptococcus
portant to note that healthy individuals often harbor onco- pyogenes was TrueCut Cas9 v2 (Catalogue # A36499, Ther-
genic translocations in B and T cells (42–44). T cells bearing moFisher). Cas9 RNP was made by incubating protein with
translocations can persist for months to years without evi- gRNA at a molar ratio of 1:1 at 25°C for 10 minutes immedi-
dence of pathogenicity (45–47). ately prior to electroporation.
Antagonism of the PD-1:PD-L1 costimulatory pathway can
result in organ-specific and systemic autoimmunity (17, 48). Lentiviral vector manufacturing
PD-1 has been reported to function as a haploinsufficient tu- The 8F TCR recognizes the HLA-A*0201 SLLMWITQC
mor suppressor in mouse T cells (49). Our patients have had epitope on NY-ESO-1 and LAGE-1. The 8F TCR was isolated
engraftment with PD-1 deficient T cells and to date, there is from a T cell clone obtained from patient after vaccination
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 6
Multiplexed genetic engineering of human hematopoietic stem and progenitor tumor-reactive lymphocytes promotes intratumoral T-cell persistence and
cells using CRISPR/Cas9 and AAV6. eLife 6, e27873 (2017). rejection of established tumors. Cancer Res. 76, 2087–2093 (2016).
doi:10.7554/eLife.27873 Medline doi:10.1158/0008-5472.CAN-15-3352 Medline
10. P. F. Robbins, R. A. Morgan, S. A. Feldman, J. C. Yang, R. M. Sherry, M. E. Dudley, 23. L. Cherkassky, A. Morello, J. Villena-Vargas, Y. Feng, D. S. Dimitrov, D. R. Jones, M.
J. R. Wunderlich, A. V. Nahvi, L. J. Helman, C. L. Mackall, U. S. Kammula, M. S. Sadelain, P. S. Adusumilli, Human CAR T cells with cell-intrinsic PD-1 checkpoint
Hughes, N. P. Restifo, M. Raffeld, C.-C. R. Lee, C. L. Levy, Y. F. Li, M. El-Gamil, S. L. blockade resist tumor-mediated inhibition. J. Clin. Invest. 126, 3130–3144 (2016).
Schwarz, C. Laurencot, S. A. Rosenberg, Tumor regression in patients with doi:10.1172/JCI83092 Medline
metastatic synovial cell sarcoma and melanoma using genetically engineered 24. J. Ren, X. Liu, C. Fang, S. Jiang, C. H. June, Y. Zhao, Multiplex genome editing to
lymphocytes reactive with NY-ESO-1. J. Clin. Oncol. 29, 917–924 (2011). generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 23,
doi:10.1200/JCO.2010.32.2537 Medline 2255–2266 (2017). doi:10.1158/1078-0432.CCR-16-1300 Medline
11. A. P. Rapoport, E. A. Stadtmauer, G. K. Binder-Scholl, O. Goloubeva, D. T. Vogl, S. 25. J. Ren, X. Zhang, X. Liu, C. Fang, S. Jiang, C. H. June, Y. Zhao, A versatile system
F. Lacey, A. Z. Badros, A. Garfall, B. Weiss, J. Finklestein, I. Kulikovskaya, S. K. for rapid multiplex genome-edited CAR T cell generation. Oncotarget 8, 17002–
Sinha, S. Kronsberg, M. Gupta, S. Bond, L. Melchiori, J. E. Brewer, A. D. Bennett, 17011 (2017). doi:10.18632/oncotarget.15218 Medline
A. B. Gerry, N. J. Pumphrey, D. Williams, H. K. Tayton-Martin, L. Ribeiro, T. Holdich, 26. X. Liu, Y. Zhang, C. Cheng, A. W. Cheng, X. Zhang, N. Li, C. Xia, X. Wei, X. Liu, H.
S. Yanovich, N. Hardy, J. Yared, N. Kerr, S. Philip, S. Westphal, D. L. Siegel, B. L. Wang, CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res. 27,
Levine, B. K. Jakobsen, M. Kalos, C. H. June, NY-ESO-1-specific TCR-engineered T 154–157 (2017). doi:10.1038/cr.2016.142 Medline
cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 27. E. K. Moon, R. Ranganathan, E. Eruslanov, S. Kim, K. Newick, S. O’Brien, A. Lo, X.
21, 914–921 (2015). doi:10.1038/nm.3910 Medline Liu, Y. Zhao, S. M. Albelda, Blockade of programmed death 1 augments the ability
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 7
100, 12871–12876 (2003). doi:10.1073/pnas.2135498100 Medline TCRs generated by phage display provide CD4+ T cells with the ability to recognize
37. M. Kosicki, K. Tomberg, A. Bradley, Repair of double-strand breaks induced by and kill tumor cell lines. J. Immunol. 179, 5845–5854 (2007).
CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat. doi:10.4049/jimmunol.179.9.5845 Medline
Biotechnol. 36, 765–771 (2018). doi:10.1038/nbt.4192 Medline 54. S. R. Ferdosi, R. Ewaisha, F. Moghadam, S. Krishna, J. G. Park, M. R. Ebrahimkhani,
38. D. Maddalo, E. Manchado, C. P. Concepcion, C. Bonetti, J. A. Vidigal, Y.-C. Han, P. S. Kiani, K. S. Anderson, Multifunctional CRISPR-Cas9 with engineered
Ogrodowski, A. Crippa, N. Rekhtman, E. de Stanchina, S. W. Lowe, A. Ventura, In immunosilenced human T cell epitopes. Nat. Commun. 10, 1842 (2019).
vivo engineering of oncogenic chromosomal rearrangements with the doi:10.1038/s41467-019-09693-x Medline
CRISPR/Cas9 system. Nature 516, 423–427 (2014). doi:10.1038/nature13902 55. M. Kalos, B. L. Levine, D. L. Porter, S. Katz, S. A. Grupp, A. Bagg, C. H. June, T cells
Medline with chimeric antigen receptors have potent antitumor effects and can establish
39. L. Poirot, B. Philip, C. Schiffer-Mannioui, D. Le Clerre, I. Chion-Sotinel, S. Derniame, memory in patients with advanced leukemia. Sci. Transl. Med. 3, 95ra73 (2011).
P. Potrel, C. Bas, L. Lemaire, R. Galetto, C. Lebuhotel, J. Eyquem, G. W.-K. Cheung, doi:10.1126/scitranslmed.3002842 Medline
A. Duclert, A. Gouble, S. Arnould, K. Peggs, M. Pule, A. M. Scharenberg, J. Smith,
Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf”
adoptive T-cell immunotherapies. Cancer Res. 75, 3853–3864 (2015). ACKNOWLEDGMENTS
doi:10.1158/0008-5472.CAN-14-3321 Medline We thank the Human Immunology Core at the University of Pennsylvania for
40. W. Qasim, H. Zhan, S. Samarasinghe, S. Adams, P. Amrolia, S. Stafford, K. Butler, providing leukocytes for research, the Clinical Cell and Vaccine Production
C. Rivat, G. Wright, K. Somana, S. Ghorashian, D. Pinner, G. Ahsan, K. Gilmour, G. Facility for GMP cell manufacturing, the Hospital of the University of
Lucchini, S. Inglott, W. Mifsud, R. Chiesa, K. S. Peggs, L. Chan, F. Farzeneh, A. J. Pennsylvania Apheresis Unit for peripheral blood mononuclear cell collections
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 8
References (51–55)
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 9
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 1. Feasibility of CRISPR-Cas9 NYCE T cell engineering. (A) Schematic representation of
CRISPR-Cas9 NYCE T cells. (B) Large-scale expansion of NYCE T cells. Autologous T cells were
transfected with Cas9 protein complexed with single guide RNAs (ribonucleoprotein; RNP complex)
against TRAC, TRBC (i.e., endogenous TCR deletion) and PDCD1 (i.e., PD-1 deletion) and
subsequently transduced with a lentiviral vector to express a transgenic NY-ESO-1 cancer-specific
TCR. Cells were expanded in dynamic culture for 8 to 12 days. On the final day of culture, NYCE
T cells were harvested and cryopreserved in infusible medium. The total number of enriched T cells
during culture is plotted for all four subjects (UPN07, UPN27, UPN35 and UPN39). (C) NY-ESO-1
TCR transduction efficiency was determined in harvested infusion products by flow cytometry. Data
are gated on live CD3-expressing and Vβ8.1 or dextramer positive lymphocytes and further gated on
CD4 and CD8 positive cells. (D) The frequencies of TRAC, TRBC and PDCD1 gene-disrupted total
cells in NYCE infusion products were measured using chip-based digital PCR. All data are
representative of at least two independent experiments. Error bars represent mean +/− SEM.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 10
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 2. Potency and immunogenicity of CRISPR-Cas9 engineered T cells. (A) Cytotoxicity of NYCE T cells co-
cultured with HLA-A*0201 positive Nalm-6 tumor cells engineered to express NY-ESO-1 and luciferase. Patient
T cells transduced with the NY-ESO-1 TCR without CRISPR-Cas9 editing (NY-ESO-1 TCR) and untransduced T cells
with CRISPR-Cas9 editing of TRAC, TRBC and PDCD1 (denoted as here as CRISPR) were included as controls (n = 4
patient T cell infusion products). Asterisks indicate statistical significance determined by paired t-tests between
groups (*P < 0.05). Error bars represent SEM. (B) Levels of soluble interferon-γ produced by patient NYCE T cell
infusion products (denoted as NYCE) following a 24-hour co-culture with anti-CD3/CD28 antibody-coated beads or
NY-ESO-1-expressing Nalm-6 target cells. Patient NY-ESO-1 TCR transduced T cells (NY-ESO-1 TCR) and
untransduced, CRISPR-Cas9 edited T cells (denoted as CRISPR) served as controls. Error bars indicate SEM.
(C) Quantification of residual Cas9 protein in NYCE T cell infusion products in clinical-scale manufacturing is shown
over time. Asterisks depict statistical significance determined by paired t-tests between time points (*P < 0.05).
(D) Results from the fluorescence-based indirect ELISA screen performed to detect antibodies against Cas9 protein
in the sera of three patients treated with NYCE T cells. Each dot represents the amount of anti-Cas9 signals detected
in patient serum prior to T cell infusion (denoted by a vertical black arrow) and at various time points following NYCE
T cell transfer. RFU = relative fluorescent units. (E) Immunoreactive Cas9-specific T cells in baseline patient
leukapheresis samples were detected. Representative flow cytometry plots (left panel) from two patients whose
T cells were positive for interferon-γ in response to Cas9 peptide stimulation. Unstimulated T cells treated with
vehicle alone (dimethyl sulfoxide, DMSO) served as a negative control, while matched T cells stimulated with phorbol
myristate acetate (denoted as PMA)/Ionomycin served as a positive control. Bar graphs (right panel) show the
frequency of ex vivo CD4+ and CD8+ T cells from patients or healthy donor controls (n = 6) that secrete interferon-γ
in response to stimulation with three different Cas9 peptide pools. The background frequency of interferon-γ-
expressing T cells (unstimulated control group, DMSO alone) is subtracted from the values shown in the bar graph.
Error bars depict SD.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 11
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 3. Sustained in vivo expansion and persistence of CRISPR-Cas9 engineered T cells in
patients. (A) The total number of vector copies per microgram of genomic DNA of the NY-ESO-1 TCR
transgene in the peripheral blood (UPN07, UPN35 and UPN39), bone morrow (UPN07 and UPN35;
multiple myeloma), and tumor (UPN39; sarcoma) is shown pre- and post-NYCE T cell infusion.
(B) Calculated absolute numbers of NY-ESO-1 TCR-expressing T cells per microliter of whole blood
from the time of infusion to various post-infusion time points in the study are shown. The limit of
detection is approximately 2.5 cells/μl of whole blood. (C) Frequencies of CRISPR-Cas9-edited
T cells (TRAC, TRBC and PDCD1 knockout) before and following adoptive cell transfer are depicted.
Error bars indicate SD.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 12
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 4. Fidelity of CRISPR-Cas9 gene editing. (A) Genomic distribution of oligonucleotide (dsODN)
incorporation sites, which mark locations of double strand breaks. The ring indicates the human
chromosomes aligned end-to-end, plus the mitochondrial chromosome (labeled M). The targeted
cleavage sites are on chromosomes 2, 7, and 14. The frequency of cleavage and subsequent dsODN
incorporation is shown on a log scale on each ring (pooled over 10 Mb windows). The purple inner
most ring plots all alignments identified. The green ring shows “pile ups” of three or more overlapping
sequences; the blue ring shows alignments extending along either strand from a common dsODN
incorporation site (“flanking pairs”); the red ring shows reads with matches to the gRNA (allowing
<6 mismatches) within 100 bp (“target matched”). (B) Distribution of inferred positions of cleavage
and dsODN incorporation at an on-target locus. Incorporations in different strand orientations are
shown on the positive (red) and negative (blue) y-axis. The percentage in the bottom right corner is
an estimate of the number of incorporations associated with the on-target site (based on pileups)
captured within the allowed window of 100 bps. (C) Sequences of sites of cleavage and dsODN
incorporation are shown, annotated by whether they are on target or off target (“Target”); the total
number of unique alignments associated with the site (“Abund”); and an identifier indicating the
nearest gene (“Gene ID”). *Symbols after the gene name indicate that the site is within the
transcription unit of the specific gene, whereas the ~ symbol indicates the gene appears on the
allOnco cancer-associated gene list.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 13
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 5. Detection of chromosomal translocations in engineered T cells following CRISPR-Cas9
gene editing. (A) Evaluation of chromosomal translocations in NYCE T cell infusion products during
the course of large-scale culture is shown. For the 12 monocentromeric translocation assays
conducted, a positive reference sample that contains 1 × 103 copies of the synthetic template plasmid
was evaluated as a control and the percent difference between expected and observed marking was
calculated. The absence of amplification from the 12 reactions that correspond to the different
chromosomal translocations indicates assay specificity (see supplemental methods).
(B) Longitudinal analysis of chromosomal translocations in vivo in three patients pre- and post-NYCE
T cell product infusion is displayed. In (A) and (B), error bars denote SD. For graphical purposes, the
proportions of affected cells were plotted on a log scale; a value of 0.001% indicates that
translocations were not detected.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 14
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 6. Single-cell RNA sequencing of patient UPN39 CRISPR-Cas9 engineered NYCE T cells pre-
and post-infusion. (A) Venn diagram showing relative numbers of NY-ESO-1 TCR-positive cells with
TRAC, TRBC and/or PDCD1 mutations in the NYCE T cell infusion product (Day 0). (B) Proportions
of pre-infusion (Day 0) and post-infusion (Days 10 and 113) T cells without mutations (wild-type),
with TRAC, TRBC, PDCD1 mutations or expressing the NY-ESO-1 TCR transgene. Numbers of cells
belonging to each of these categories are listed below the graph. (C) Analysis of NY-ESO-1 TCR-
positive (right) and TCR–negative (left) cells without mutations (wild-type) or with single, double or
triple mutations at Day 0 (NYCE T cell infusion product) and Day 113 post-NYCE T cell infusion.
(D) UMAP plots of gene expression data. Analysis was performed on all T cells integrated across time
points, but only NY-ESO-1 TCR-expressing cells, split by time point, are shown (top panel). The
increase in TCF7 expression is indicative of an acquired central memory phenotype (bottom panel,
same cells). (E) Heat map showing scaled expression of differentially expressed genes in NY-ESO-1
TCR-positive T cells across time points. Color scheme is based on scaled gene expression from –2
(yellow) to 2 (purple).
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 15
Downloaded from http://science.sciencemag.org/ on February 6, 2020
Fig. 7. Clinical responses and patient outcome following infusion of CRISPR-Cas9 engineered
NYCE T cells. (A) Swimmer’s plot describing time on study for each patient, duration of follow-up off
study (defined as survival beyond progression or initiation of other cancer therapy) and present
status (differentially colored) is shown. Arrows indicate ongoing survival. (B) Changes in kappa light
chain levels (mg/L × 103) in patient UPN07 following NYCE T cell product infusion are depicted.
Vertical black arrow indicates initiation of a D-ACE salvage chemotherapy regimen (defined as
intravenous infusion of cisplatin, etoposide, cytarabine and dexamethasone). (C) Longitudinal M-
Spike levels (g/dL) in patient UPN35 post-NYCE T cell product administration are shown. Vertical
black arrows denote administration of combination therapy with elotuzumab, pomalidomide and
dexamethasone. (D) Computed tomography scans demonstrating tumor regression in patient
UPN39 following administration of an autologous NYCE T cell infusion product. Radiologic studies
were obtained before therapy and after adoptive transfer of NYCE T cells. Tumor is indicated by red
X. SD, stable disease; PD, progressive disease. (E) Cytolytic capacity of NY-ESO-1-specific CD8+
T cells recovered at the indicated month after infusion and expanded from patients is shown. PBMC
samples collected after NYCE T cell product infusion were expanded in vitro in the presence of
NY-ESO-1 peptide and IL-2. The ability of expanded effector cells to recognize antigen and elicit
cytotoxicity was tested in a 4-hour 51CR release assay incorporating Nalm-6 NY-ESO-1+, parental
Nalm-6 (NY-ESO-1-) and A375 melanoma cells (NY-ESO-1+). All target cell lines were HLA-A*02
positive. Assays were performed in triplicate and error bars represent SD.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 16
Table 1. Patient demographics and date of engineered T cell infusion. UPN, unique patient number; MM, multiple
myeloma; BM, bone marrow; XRT, radiation therapy; ASCT, autologous hematopoietic stem cell transplant; ND, not
done.
Subject ID
Prior LAGE-1* /
(UPN) / Gender / Clinical
Diagnosis Prior Therapy Transplant / NY-ESO-1* /
Infusion Age Sites
Surgery NY-ESO-1**
date
Lenalidomide,
pomalidomide,
bortezomib,
carfilzomib,
UPN35 Female / IgG kappa BM, lytic
daratumumab, ASCT x 3 Pos/Pos/Neg
1/7/19 66 years MM 2008 bone lesions
panobinostat
Doxorubicin, Resection/
Myxoid/ ifosfamide, debulking × 2,
Abdominal/
UPN39 Male / round cell XRT 60-Gy, left nephrec-
pelvic ND/ND/Pos
3/18/19 66 years liposarcoma trabectedin, tomy/ partial
masses
2012 gemcitabine, sigmoid
taxol, XRT resection
Lenalidomide,
pomalidomide,
bortezomib,
carfilzomib,
Kappa light
UPN07 Female / BM, lytic daratumumab,
chain MM ASCT x 2 Pos/Pos/Neg
8/5/19 62 years bone lesions anti-CD38
2009
immunoconjugate
(6 lines; see
supplementary
materials)
*qPCR
**Immunohistochemistry
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 17
Table 2. List of adverse events on the study.
All
AE Category Toxicity Grade 1/2 Grade 3/4
Grades
Anemia 2 1 1
Leukopenia 4 - 4
Hematologic Neutropenia 4 1 3
Thrombocytopenia 6 3 3
Lymphopenia 1 - 1
Upper Respiratory 1 1 -
Infection
Febrile Neutropenia 2 - 2
Hypercalcemia 1 1 -
Dysgeusia 1 1 -
Headache 1 1 -
Neurologic Paresthesia 2 2 -
Syncope 1 - 1
Pain 3 3 -
Aspiration 1 - 1
Respiratory Nasal congestion 1 1 -
Cough 2 2 -
Lower GI bleed 1 1 -
Gastrointestinal
Vomiting 1 1 -
Alopecia 1 1 -
Other Phlebitis 1 1 -
LE edema 1 1 -
Total 50 30 20
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 18
Table 3. iGUIDE measurement of on-target editing efficiency for each gene by final product.
First release: 6 February 2020 www.sciencemag.org (Page numbers not final at time of first release) 19
CRISPR-engineered T cells in patients with refractory cancer
Edward A. Stadtmauer, Joseph A. Fraietta, Megan M. Davis, Adam D. Cohen, Kristy L. Weber, Eric Lancaster, Patricia A. Mangan,
Irina Kulikovskaya, Minnal Gupta, Fang Chen, Lifeng Tian, Vanessa E. Gonzalez, Jun Xu, In-young Jung, J. Joseph Melenhorst,
Gabriela Plesa, Joanne Shea, Tina Matlawski, Amanda Cervini, Avery L. Gaymon, Stephanie Desjardins, Anne Lamontagne,
January Salas-Mckee, Andrew Fesnak, Donald L. Siegel, Bruce L. Levine, Julie K. Jadlowsky, Regina M. Young, Anne Chew,
Wei-Ting Hwang, Elizabeth O. Hexner, Beatriz M. Carreno, Christopher L. Nobles, Frederic D. Bushman, Kevin R. Parker, Yanyan
Qi, Ansuman T. Satpathy, Howard Y. Chang, Yangbing Zhao, Simon F. Lacey and Carl H. June
SUPPLEMENTARY http://science.sciencemag.org/content/suppl/2020/02/05/science.aba7365.DC1
MATERIALS
RELATED http://science.sciencemag.org/content/sci/367/6478/616.full
CONTENT
http://science.sciencemag.org/content/sci/early/2020/02/05/science.aba9844.full
REFERENCES This article cites 55 articles, 20 of which you can access for free
http://science.sciencemag.org/content/early/2020/02/05/science.aba7365#BIBL
PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions
Science (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement of
Science, 1200 New York Avenue NW, Washington, DC 20005. The title Science is a registered trademark of AAAS.
Copyright © 2020, American Association for the Advancement of Science