You are on page 1of 18

nature reviews clinical oncology https://doi.org/10.

1038/s41571-023-00729-2

Review article Check for updates

Advancing CAR T cell


therapy through the use of
multidimensional omics data
Jingwen Yang1,6, Yamei Chen , Ying Jing
1,6
, Michael R. Green
2,6 3,4
& Leng Han 1,5

Abstract Sections

Despite the notable success of chimeric antigen receptor (CAR) T cell Introduction

therapies in the treatment of certain haematological malignancies, Multidimensional profiling


challenges remain in optimizing CAR designs and cell products, technologies

improving response rates, extending the durability of remissions, Target discovery for CAR T cell
therapy
reducing toxicity and broadening the utility of this therapeutic
modality to other cancer types. Data from multidimensional omics Understanding and enhancing
CAR T cell efficacy and
analyses, including genomics, epigenomics, transcriptomics, T cell persistence
receptor-repertoire profiling, proteomics, metabolomics and/or Minimizing CAR T cell-related
microbiomics, provide unique opportunities to dissect the complex toxicities
and dynamic multifactorial phenotypes, processes and responses of Conclusions
CAR T cells as well as to discover novel tumour targets and pathways
of resistance. In this Review, we summarize the multidimensional
cellular and molecular profiling technologies that have been used
to advance our mechanistic understanding of CAR T cell therapies.
In addition, we discuss current applications and potential strategies
leveraging multi-omics data to identify optimal target antigens and
other molecular features that could be exploited to enhance the
antitumour activity and minimize the toxicity of CAR T cell therapy.
Indeed, fully utilizing multi-omics data will provide new insights into
the biology of CAR T cell therapy, further accelerate the development
of products with improved efficacy and safety profiles, and enable
clinicians to better predict and monitor patient responses.
1
Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University,
Houston, TX, USA. 2Department of Biochemistry and Molecular Biology, The University of Texas Health Science
Center at Houston McGovern Medical School, Houston, TX, USA. 3Department of Lymphoma and Myeloma,
The University of Texas MD Anderson Cancer Center, Houston, TX, USA. 4Department of Genomic Medicine,
The University of Texas MD Anderson Cancer Center, Houston, TX, USA. 5Department of Translational Medical
Sciences, College of Medicine, Texas A&M University, Houston, TX, USA. 6These authors contributed equally:
Jingwen Yang, Yamei Chen, Ying Jing. e-mail: MGreen5@mdanderson.org; leng.han@tamu.edu

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 211


Review article

Key points fields22,23, including CAR T cell therapy24–26. CAR T cells are living cells
that can actively sense and respond to a wide variety of extrinsic and
intrinsic factors27. Therefore, identification of detrimental phenotype
•• Multidimensional omics data, encompassing genomics, epigenomics, heterogeneity and pathway regulation of pre-manufacturing autolo-
transcriptomics, T cell receptor-repertoire profiling, proteomics, gous T cells through bulk and single-cell profiling28–30 might contribute
metabolomics and microbiomics, have been exploited to advance our to the quality control of CAR T cell products or inform strategies for
mechanistic understanding of chimeric antigen receptor (CAR) T cell CAR design or product manufacture that improve their functionality.
therapy. Moreover, the use of multi-omics strategies to monitor the temporally
dynamic expansion of functional and/or dysfunctional CAR T cell sub-
•• Utilization of multi-omics data derived from both tumour and non- populations24,31, characterize CAR T cell transcriptomic32, epigenomic33
malignant tissues at the bulk and/or single-cell levels is a powerful and metabolic34 dynamics, delineate tumour microenvironment (TME)
approach to identifying optimal targets for highly efficacious and safe features that restrict CAR T cell infiltration and suppress CAR T cell
CAR T cell therapy. cytotoxicity35, determine the origins and functional consequences of
key cytokines36,37, and predict on-target, off-tumour toxicities in non-
•• Integration of bulk and/or single-cell multidimensional omics malignant tissues38 could facilitate biomarker discovery and improve
data has been applied to investigate key determinants of CAR T cell our mechanistic understanding of CAR T cell therapy responses and
persistence and antitumour efficacy, including T cell states and toxicities.
phenotypes, tumour cell characteristics, the tumour microenvironment In this Review, we highlight the potential of multi-omics data
and the microbiome. to advance CAR T cell therapy. We first provide an overview of the
multidimensional profiling technologies that have been utilized in
•• Leveraging multi-omics data promises to elucidate the mechanisms the investigation of CAR T cell therapy. Next, we discuss strategies
underlying CAR T cell-related toxicities, including cytokine-release leveraging multi-omics data to discover single and combinatorial
syndrome, immune effector cell-associated neurotoxicity syndrome CAR targets. We further discuss molecular features and other key fac-
and on-target, off-tumour toxicities. tors that are associated with enhanced antitumour responses and/or
decreased toxicity of CAR T cell therapy as uncovered by multi-omics
profiling. Exploiting current and emerging multidimensional
Introduction omics profiling technologies will enable a comprehensive understand-
Chimeric antigen receptors (CARs) are synthetic fusion receptors ing of the molecular biology of CAR T cell therapies, ultimately enabling
designed to redirect T cells to recognize and eliminate cancer cells that the clinical benefits of these treatments to be maximized.
express cognate antigens1. To achieve this aim, CARs have typically been
constructed using antibody-based moieties targeting specific tumour- Multidimensional profiling technologies
associated antigens, which are linked via a hinge and transmembrane Data from genomics, epigenomics, transcriptomics, TCR-repertoire
domain to intracellular signalling motifs derived from both the T cell profiling, proteomics, metabolomics and microbiomics studies have
receptor (TCR) CD3ζ chain and a co-stimulatory receptor (such as CD28 been applied to address the remaining key challenges for CAR T cell
and/or 4-1BB)1. Transgenes encoding these constructs are introduced therapy. Leveraging such multi-omics data can provide novel insights
into autologous (patient-derived) or allogeneic (healthy donor- into tumour and non-malignant cell characteristics, cell state tran-
derived) T cells ex vivo, followed by selection and in vitro expansion sitions, cell types, and cell–cell interactions in the TME that affect
of CAR-expressing T cells before they are infused into the patient. treatment outcomes of CAR T cell therapy (Fig. 1 and Table 1).
CAR T cell therapy has moved to the forefront of therapy for relapsed
and/or refractory B cell malignancies, owing to impressive response Genomics
rates and durability in patients with B cell acute lymphoblastic leukae- Comparisons of DNA sequencing (DNA-seq) data from tumour and non-
mia (B-ALL)2, large B cell lymphoma (LBCL)3, mantle cell lymphoma4, malignant tissue samples have identified numerous tumour-associated
follicular lymphoma5 or multiple myeloma6. In addition, CAR T cells somatic mutations, some of which might generate tumour-specific
have been demonstrated to induce clinical responses in patients with neoantigens that are presented on the cell surface and could potentially
various advanced-stage solid tumours, including glioblastoma7, pancre- serve as novel targets for CARs or TCR-like constructs39,40. To date,
atic ductal adenocarcinoma8, sarcoma9, gastrointestinal cancers10 and however, attempts to target cancer cells with neoantigen-directed CAR
castration-resistant prostate cancer11, albeit less frequently and more T cells have been limited to a few preclinical and clinical studies41–43.
transiently than in patients with B cell malignancies. Nevertheless, the Alternatively, tumour-associated mutations and complex genomics
clinical success of CAR T cell therapy faces several challenges, includ- alterations might influence clinical responses to CAR T cell therapy44–47.
ing the paucity of robustly expressed tumour-specific antigens in Genome-wide pooled CRISPR–Cas9 knockout library screening fol-
many malignancies12,13, CAR T cell exhaustion14,15, limited CAR T cell lowed by sequencing-based quantification of the frequencies of single
persistence16,17 and potential life-threatening toxicities18,19. guide RNAs enables large-scale, high-throughput studies to assess the
Multi-omics data, encompassing genomics, epigenomics, tran- influence of individual genes under different conditions and thereby
scriptomics, proteomics and beyond, can provide deep insights into identify cellular functional dependencies48,49. Such functional genom-
the abundance and/or variation of biological molecules across multiple ics profiling has resulted in the identification of key genes involved in
dimensions in various tissues or cells20,21. The rapid development of T cell activation50, persistence51, cytotoxicity52–54 and dysfunction55
multidimensional profiling strategies and approaches for their integra- as well as genetic alterations in tumour cells that influence resist-
tion has facilitated the use of multi-omics data to characterize the com- ance to treatment56, highlighting regulators that could potentially be
plex molecular features of diverse cellular processes in broad research exploited to enhance the efficacy of CAR T cell therapy. Meanwhile,

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 212


Review article

Sample Multi-omics Features Clinical applications

CAR design Identify targets


Genome
Recognition

Tumour cells

Me Me Ac Me Epigenome Enhance efficacy


Bulk
CAR T cell

Non-malignant cells

Transcriptome Cell functional phenotype


Cytotoxic

TCR repertoire

Memory Reduce toxicity


Single cell
P P

Proteome CAR T cell

Exhausted
Cytokine storm and
Tumour microenvironment cytokine-release
syndrome
Spatial Metabolome
Immunosuppression

Microbiome On-target,
Cellular composition and off-tumour
cell–cell interactions Neurotoxicities toxicities

Fig. 1 | Overview of applications of multi-omics data in CAR T cell therapy. chimeric antigen receptor (CAR) T cell therapy. The valuable multi-omics data
Several types of samples, including bulk tissue specimens, single-cell suspensions facilitates the characterization of diverse tumour and non-malignant cell features,
or spatially preserved tissue slides (left panel), can be used for multidimensional cell functional phenotypes (for example, of CAR T cells both prior to and after
omics analyses, including genomics, epigenomics, transcriptomics, T cell infusion), and various properties of the tumour microenvironment (middle right
receptor (TCR)-repertoire, proteomics, metabolomics and/or microbiomics panel), which will enable advances in the safety and efficacy of CAR T cell therapy
profiling (middle left panel), across a multitude of applications in the field of through improvements in target identification and cell engineering (right panel).

pooled CRISPR-based gain-of-function screening in combination with Combination analyses with assay for transposase-accessible chro-
single-cell RNA sequencing (scRNA-seq) has been used to measure the matin using sequencing (ATAC-seq) and RNA sequencing (RNA-seq)
abundance and assess the cellular states of T cells expressing different can facilitate the identification of epigenomics regulatory modules
knock-in constructs and highlighted a novel chimeric cytokine– that control cellular programmes related to CAR T cell exhaustion and
co-stimulatory receptor for CAR T cells that improves solid tumour cytotoxicity32,66–69, thus providing a multitude of insights into potential
clearance57. In summary, functional genomics profiling is a powerful epigenetic reprogramming strategies to improve efficacy.
method to elucidate mechanistic regulators of CAR T cell functions Advances in single-cell epigenomics profiling70 have enabled eval-
that might ultimately be leveraged to refine CAR design or cellular uation of the epigenetic regulatory landscape within individual CAR
engineering. T cells71. Combining data derived using single-cell ATAC-seq with tran-
scriptomics and proteomics data has helped to identify the patterns
Epigenomics of epigenetic regulation associated with functional subpopulations of
Epigenetic factors, including DNA methylation, histone modifications, CAR T cells28. Collectively, the emerging evidence from epigenomics
and chromatin accessibility and 3D architecture, enable the integration profiling studies demonstrates the importance of dynamic epigenetic
of multiple cellular signals to dynamically regulate the gene expression alterations within CAR T cells in determining the molecular features
programme and, ultimately, the phenotype and function of T cells58,59. associated with therapeutic efficacy.
Epigenetic reprogramming of CAR T cells through perturbation of
epigenetic modulators therefore has the potential to boost antitu- Transcriptomics
mour efficacy60–64. Genome-wide epigenomics profiling techniques RNA-seq has an essential role in understanding cellular states and has
have become the optimal approach for investigating epigenetic enabled tremendous progress in the characterization of tumour hetero­
regulatory landscapes65, including those of CAR T cells. For example, geneity and the TME72–74. In the field of CAR T cell therapy, RNA-seq is
DNA methylation array analyses can help to identify differentially most commonly applied for differentially expressed gene analysis
methylated sites associated with response to CAR T cell therapy33,66. under various conditions to delineate the cell-state transitions that

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 213


Review article

Table 1 | Representative multi-omics profiling approaches applied to study CAR T cell therapy

Omics approach Profiling technology Examples of insights obtained

Genomics DNA-seq Identification of genomics alterations as potential targets or as biomarkers for therapeutic outcome44–46
CRISPR screening Defining dependencies for the cytotoxic activity of CAR T cells and determinants of the sensitivity of
cancer cells to CAR T cell-mediated cytotoxicity52,53
Epigenomics DNA methylation arrays Definition of differentially methylated genomic loci related to CAR T cell functions and therapeutic
responses33,66
ATAC-seq Characterization of changes in accessible chromatin regions of genes that affect CAR T cell
functions32,68
ChIP-seq Identification of transcriptional reprogramming of the exhaustion-associated epigenome in exhausted
CAR T cells63
scATAC-seq Discovery of epigenetic differences correlated with distinct CAR T cell subpopulations28,71
Transcriptomics RNA-seq Identification of transcriptional changes related to CAR T cell functions32,76; discovery of novel targets
based on differential expression and RNA dysregulation in malignant versus non-malignant tissues80,82
scRNA-seq Characterization of CAR T cell functional states that are associated with particular cellular conditions,
CAR designs/cell products, clinical responses and toxicities87; discovery of subpopulations of non-
malignant and tumour cells that cause toxicities and disease relapse, respectively90,203; identification of
hostile factors in the tumour microenvironment that affect CAR T cell function and, therefore, hamper
solid tumour elimination91
TCR-repertoire profiling TCR-seq Kinetic profiling of different TCR clonotypes throughout the treatment process96,97
Proteomics Mass spectrometry Information on the surfaceome (expression landscape of cell-surface proteins) and immunopeptidome
(repertoire of MHC-presented peptide epitopes) as a resource for CAR target identification100,103,106;
phosphoproteomics characterization to evaluate CAR downstream signalling109
Mass cytometry Identification of immune markers for monitoring of CAR T cell activation, proliferation and exhaustion114
Metabolomics Mass spectrometry Discovery of metabolic reprogramming in CAR T cells, which could enhance antitumour efficacy34,124,125
Metabolic flux analysis Assessment of dynamic metabolic pathway activity related to different cell conditions129
Mass cytometry of metabolic Characterization of the metabolic adaptation of CAR T cell products after infusion242
proteins
Microbiomics 16S rRNA-seq and Identification of features of the gut microbiota correlated with response and toxicities141
metagenomics shotgun
sequencing
16S rRNA-seq, 16S ribosomal RNA sequencing; ATAC-seq, assay for transposase-accessible chromatin with high-throughput sequencing; CAR, chimeric antigen receptor; ChIP-seq, chromatin
immunoprecipitation sequencing; DNA-seq, DNA sequencing; RNA-seq, RNA sequencing; scATAC-seq, single-cell ATAC-seq; scRNA-seq, single-cell RNA-seq; TCR, T cell receptor; TCR-seq,
TCR sequencing.

are associated with efficacy15,32,75,76. Moreover, monitoring changes in memory, exhaustion and other cellular programmes can be affected by
the transcription of genes encoding various cytokines, which are key variations in CAR T cell conditions or designs52,76,87,88 and are associated
mediators of CAR T cell activity, is also crucial to improve efficacy and with clinical response and toxicity15. Beyond profiling of the CAR T cell
reduce the risk of cytokine-release syndrome (CRS), immune effec- products, applying scRNA-seq to non-malignant tissues and tumours
tor cell-associated neurotoxicity syndrome (ICANS) and other toxici- can help to identify rare cell subpopulations in order to elucidate the
ties67,77–79. Application of total RNA-seq provides additional information, mechanisms underlying treatment-related toxicities15,89,90. Addition-
for example, on the expression of non-coding RNAs that can regulate ally, scRNA-seq can provide rich gene expression data across various
immunomodulatory molecules and thereby affect CAR T cell cyto- cell types and is therefore an optimal unbiased approach for exploring
toxicity80. Furthermore, dysregulation of post-transcriptional RNA factors in the TME that might affect the antitumour activity of CAR
processing can lead to variant transcripts, thus potentially generating T cells91,92. Such insights could offer potential opportunities to over-
different protein isoforms that enable escape from CAR recognition81. come immunosuppressive features of the TME and seek solutions for
Therefore, increasing interest is being placed on exploiting the existing the expansion of CAR T cell applications in solid tumours91,93.
and expanding tumour RNA-seq data to discover novel CAR targets
derived from the differential expression and dysregulation of RNAs82,83. TCR-repertoire profiling
scRNA-seq, which generates data on the transcriptome of indi- The TCR is a heterodimeric cell-surface protein complex comprising
vidual cells, has become a well-established method since 2009 (ref. 84) heterogeneous TCRα and TCRβ or TCRγ and TCRδ chains generated
and is widely applied in cancer research85,86. Notably, scRNA-seq is an through random genetic recombination of different variable, diversity,
emerging method of investigating features of CAR T cells in preclini- junctional and constant gene segments in individual T cell clones; this
cal and translational research. For example, scRNA-seq enables gene process results in a varied T cell population with a highly diverse TCR
expression-based cell state examination of CAR T cell infusion prod- repertoire to mediate antigen-specific adaptive immune responses94.
ucts, with data demonstrating that signatures of cytotoxicity, central High-throughput technologies have been developed to sequence the

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 214


Review article

TCR chains expressed in each T cell and thus to characterize the TCR high-dimensional cellular phenotyping. Mass cytometry analysis of
repertoire and clonotypic diversity of the T cell population. Performing protein expression has been utilized for functional characterization
TCR sequencing in combination with other omics approaches, such as of CAR T cells in preclinical and clinical settings, facilitating rapid
transcriptomics profiling, enables specific TCR clonotypes to be linked monitoring of the dynamic features of these cells114–116.
with varying T cell phenotypes95. This combined analysis can help to Technologies enabling single-cell secretome analyses have also
characterize T cell dynamics through tracing of T cell clonality among been developed. For example, single-cell microchamber, proteomics
different subpopulations over time95, and such studies have demon- barcode and multiplex immunoassay chips have been used to quan-
strated the cytotoxic and proliferative features of highly expanded CAR tify the secretion of ~32 selected effector and immunostimulatory
T cell clonotypes in patients with cancer96,97. Moreover, advances in molecules by pre-infusion CAR T cell products as a measure of their
single-cell technologies have enabled simultaneous TCR sequencing and ‘polyfunctionality’, which is associated with clinical response and
scRNA-seq in individual cells, which assigns a specific TCR clonotype to toxicities117,118.
an individual T cell with a distinct transcriptional phenotype and could
thus enable TCR clonotypes to be used as a surrogate for the expansion Metabolomics
and persistence of T cell functional states throughout therapy15. With Metabolomics profiling catalogues a suite of small molecules produced
the caveat that individual TCR clonotypes can encompass a mixture during metabolic processes, providing a functional readout of various
of cell states along a functional continuum, these profiling methods cellular activities119. T cell function is regulated by intrinsic and extrinsic
offer a route to further explore whether specific clonotypically defined metabolic factors, and robust intrinsic metabolic activity is necessary
T cell states are associated with CAR T cell performance or toxicities. for effective T cell cytotoxicity120. Disrupted metabolism in the TME,
for example, owing to hypoxia and/or decreased nutrient availability,
Proteomics can lead to unmet metabolic demands in infiltrating T cells and thus
Proteomics profiling enables the identification and quantification of limit their antitumour activity16. Therefore, maintaining the metabolic
proteins and their post-translational modifications (such as phospho- fitness of CAR T cells is important to improve their therapeutic efficacy,
rylation and glycosylation) to characterize cellular pathway activity and and especially to enhance their function in the immunosuppressive
cell functions. For example, emerging proteomics profiling approaches TME of solid tumours121–123.
in the field of immuno-oncology have substantially improved our Mass spectrometry-based metabolomics enables both targeted
understanding of the mechanisms of tumour immune evasion98. Nota- metabolite quantification and comprehensive untargeted metabolite
bly, applying single-cell proteomics profiling in the context of cancer profiling. Several studies have profiled metabolites and their interme-
immunotherapy has shown early success in uncovering the clinical diates generated through various biochemical processes of interest in
relevance of distinct immune cell phenotypes in the TME93. T cells. Targeted quantification of metabolites involved in glycolysis
Traditional proteomics profiling technologies, such as protein and the tricarboxylic acid cycle has highlighted impaired intrinsic
microarrays that provide data on the expression of select proteins, metabolic processes that affect the proliferation and exhaustion of
have been applied to quantify plasma cytokine levels in patients tumour-infiltrating T cells, presenting opportunities to enhance CAR
with lymphoma or leukaemia following treatment with CAR T cells99. T cell efficacy through metabolic reprogramming124,125. Metabolomics
Advances in mass spectrometry-based proteomics techniques have data can also be leveraged for metabolic reprogramming of CAR
enabled robust high-throughput profiling of the membrane proteome, T cells to enhance their fitness and resistance to unfavourable extrinsic,
including the cell surfaceome100 and immunopeptidome101, as well as microenvironmental factors34,121,126.
signalling pathway dynamics reflected in the phosphoproteome102. Cell metabolism is variable and adaptable to environmental con-
The tumour surfaceome has been a source of novel therapeutic targets straints. Fluxomics describes the production and consumption rates
in various haematological and solid malignancies103–105. The immun- of metabolites involved in intracellular metabolic networks, which
opeptidome and surfaceome of non-malignant tissues might serve as reflect the dynamic metabolic processes within the cell127. A combina-
a reference database for the selection of tumour-specific targets and tion of stable isotopes and mass spectrometry can be used to trace
thus avoid on-target, off-tumour toxicities100,106. Phosphoproteomics and quantify dynamic metabolic fluxes, referred to as metabolic flux
can be utilized to characterize CAR downstream signalling activity analysis, thereby enabling fluxomics profiling of metabolic pathways
to evaluate the functional states associated with different CAR con- of interest such as glycolytic and mitochondrial respiration128. This
structs107–109. Thus, proteomics profiling has deepened our knowledge approach has advantages for the dynamic assessment of metabolic
of various aspects of CAR T cell therapy, especially CAR target discovery processes in CAR T cells with differing CAR designs or under different
and CAR function110. environmental conditions, with emerging fluxomics data emphasizing
Flow cytometry is the gold standard approach for both membrane the pivotal role of maintaining the metabolic fitness of CAR T cells for
and intracellular protein detection at the single-cell level for immune effective treatment123,129,130.
cell classification111. Advances in full-spectrum flow cytometry, whereby
highly sensitive detector arrays are used to capture the full emission Microbiomics
spectrum of a diverse range of fluorescent molecules, have improved The microbiota is emerging as a key factor regulating cancer progres-
throughput by increasing the number of cellular parameters that sion and therapeutic responses131,132. Several studies have demonstrated
can be evaluated in an experimental run, thus presenting a power- correlations between the gut microbiome or dietary interventions and
ful approach for protein expression analysis in studies of immuno- clinical responses to immunotherapies, in particular, immune-check-
therapy112. Mass cytometry, or cytometry by time of flight (CyTOF), point inhibitors targeting CTLA4, PD-1 or PD-L1 (refs. 133–136). Moreover,
combines flow cytometry and mass spectrometry techniques to enable microbial intervention through faecal microbial transplantation of
high-throughput, single-cell proteomics profiling (through simultane- favourable microbiota from healthy donors or responders can improve
ous evaluation of >40 proteins in millions of cells during one run)113 for the efficacy of immune-checkpoint inhibitors134,137.

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 215


Review article

Two main methods have been developed for the profiling of deconvolution algorithms, provided that the underlying reference
microbiota: 16S ribosomal RNA-seq (16S rRNA-seq)138 and metagen- signatures accurately reflect the transcriptional profiles of each cell
omics shotgun sequencing139,140. 16S rRNA-seq is more cost-effective type and pathological states within the relevant microenvironment156.
for large-scale analyses of microbial taxonomics composition but Moreover, the revolutionary applications of high-resolution single-cell
does not provide information on viral or eukaryotic components138. and/or spatial multi-omics profiling technologies have provided a
Metagenomics shotgun sequencing comprehensively covers all micro- variety of rich data on CAR T cell therapy, although these bring a level
organisms present in the sample, increasing the potential to discover of computational complexity that requires the development and
novel species with potential roles in cancer, and provides data at gene- optimization of innovative bioinformatic tools.
level resolution for analyses of functional variations, although these Second, appropriately adapting analytical methods to new appli-
benefits come at the cost of computational challenges. Microbiom- cations is a challenging yet crucial step towards achieving robust and
ics profiling of faecal samples from patients receiving CAR T cells reproducible results for biological interpretation; careful evaluation
has revealed correlations between gut microbiota composition and and correction for batch effects caused by differences in sample-
response to therapy141, suggesting promising opportunities to exploit processing protocols and data-generating platforms is required for
links with the microbiota to improve CAR T cell therapy. The current most types of omics data157. Additionally, for single-cell and spatial
studies have mainly focused on the effects of the gut microbiota, yet omics data, sparsity of data and background noise are common
the intratumour microbiota might also have an important influence challenges that necessitate the use of appropriate computational
on the antitumour immune response132,142, highlighting an additional algorithms for signal detection143,158.
dimension for future exploration. Third, the kinetics of molecular fluctuations in cells might require
further omics studies with a temporally resolved design159. Indeed,
Incorporation of spatial contexture into omics profiling several studies have demonstrated the importance of monitoring
Advances in spatial molecular profiling methods, especially spatial multi-omics variations at different stages of CAR T cell therapy to better
transcriptomics and proteomics, have enabled the large-scale and high- profile treatment-related biological changes96,97,160.
resolution characterization of localized variations in gene and protein Finally, with the accumulating volume of multi-omics data, opti-
expression using tissue sections143. Spatial transcriptomics is an emerg- mized computational approaches to integrate data from multiple
ing technology that enables high-plex or transcriptome-wide quantifi- omics modalities are desperately needed. Pioneering studies have
cation of RNAs at near-cellular144 or subcellular145 resolution that, when demonstrated the ability of a multi-omics machine learning model
integrated with non-spatially resolved single-cell transcriptomics, might to predict treatment response161, suggesting the promise of artificial
reinforce analyses of cell–cell interactions through the precise location intelligence algorithms for data integration in future studies; however,
of cells within the tissue architecture146. Similar approaches are being challenges relating to the complicated analytical procedures and to the
developed for spatial epigenomics profiling, such as spatial ATAC-seq biological translation of results remain to be addressed162–164.
and spatial cleavage under targets and tagmentation (CUT&Tag-seq),
which has proven utility in characterizing epigenetic regulation in Target discovery for CAR T cell therapy
tissues at high resolution147,148. Established technologies for spatial Identifying CAR targets with high levels of tumour specificity and cover-
proteomics, such as imaging mass cytometry, enable the simultaneous age is crucial to ensure the antitumour efficacy and reduce the toxicity
quantification and localization of ~40 proteins on a tissue slide, with of CAR T cell therapy16,165,166. In general, the ideal CAR target should be
preservation of the tissue architecture149, to deconstruct the cellular specifically and highly expressed on the surfaces of all tumour cells but
composition of the TME and explore the cytokine milieu at subcellular not on essential non-malignant cells. However, owing to shared antigen
resolution across various cancer types150–153. Furthermore, to explore expression between tumour cells and non-malignant cells as well as
the spatial organization of T cell metabolic programmes in the TME, highly heterogeneous expression among tumour cells, the identifica-
a study has utilized CyTOF to quantify the expression of proteins tion of suitable CAR T cell targets has been challenging, particularly
involved in the metabolic regulome in an individual cell and coupled for solid tumours13. In this context, the utilization and integration of
CyTOF to multiplexed ion beam imaging by time of flight (MIBI-TOF) to multidimensional omics data is a promising approach to identifying
characterize the distribution and expression of these proteins in tissue154. optimal CAR targets.
Spatial fluxomics has also been developed for dynamic metabolic profil-
ing for cancer research155. These approaches have not yet been applied Discovery of single CAR targets
to CAR T cell research, but the incorporation of spatial contexture into Transcriptomics and/or proteomics data from analyses of both malig-
omics profiling holds great promise to increase our understanding of nant and non-malignant tissues have been widely applied to discover
how cell–cell interactions and cellular neighbourhoods influence crucial CAR T cell targets that are overexpressed in tumours (Fig. 2). In a study
cellular programmes related to response and toxicity. using a novel RNA-seq-based pipeline, glypican 2 was identified as a tar-
get selectively expressed at high levels on the surface of neuroblastoma
Challenges in multi-omics data processing cells without being appreciably detected in non-malignant tissues167.
Despite the broad applications and remarkable progress of multi-omics An analysis of data from non-malignant tissues included in the Human
profiling in the realm of CAR T cell therapy, notable challenges remain Protein Atlas and the Genotype-Tissue Expression (GTEx) project and
with regard to data analysis and interpretation. First, multi-omics from tumour samples included in the Human Protein Atlas pathology
technologies using bulk tissues have broad utility in large-scale data database and the database for Differentially Expressed Proteins in
generation, but the potential masking of positive signals and averag- Cancer, encompassing 78 different tissues, 124 cell types and 20 can-
ing of signals in the context of mixed cell populations might reduce cer types, revealed the expression landscape of >100 candidate CAR
the potential for novel discoveries and the reproducibility of results. targets38. In another study, genomics, transcriptomics and immun-
This concern could potentially be addressed using newly developed opeptidomics data were integrated to identify MHC-binding peptides

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 216


Review article

Inactive
Active CAR T cell
CAR T cell

Mutant Normal
epitope peptide

Single targets
Genomics

(neoepitope) epitope

Antigen Antigen Mutant antigen Wild-type


positive negative (neoantigen) antigen
Non-malignant
Tumour tissue tissue Cell-surface antigen MHC-presented antigen
Transcriptomics

Overexpressed antigen
Proteomics

Non-malignant Neoantigen
tissues

Combinatorial targets
Tumour tissue

A B A B A B A B

"AND" logic gating "AND-NOT" logic gating

Fig. 2 | Applications of multi-omics data in CAR target identification. approaches predicated on single targets, including antigens overexpressed
Comparative analyses of genomics, transcriptomics and/or proteomics data on tumour cells and tumour cell-specific neoantigens or MHC-presented
from tumour and non-malignant tissue samples can facilitate the discovery neoepitopes; the bottom panels depict strategies based on combinatorial CAR
of novel targets and optimization of targeting strategies for chimeric antigen targets using “AND” or “AND-NOT” logic gates.
receptor (CAR) T cell therapies. The top panels illustrate CAR T cell targeting

derived from intracellular oncoproteins highly expressed in neuroblas- Neoantigens are novel tumour-specific proteins, generated
tomas; a non-mutated peptide (QYNPIRTTF) encoded by PHOX2B and mainly by somatic mutations in the tumour cells, which might consti-
presented by HLA-A*24:02 was selected as a lead target for the genera- tute ideal targets for immunotherapy39,40. To screen for neoantigens,
tion of highly specific peptide-centric CARs, which induced complete comparative whole-exome or whole-genome sequencing of matched
tumour regression in xenograft models168. Thus, bulk transcriptomics tumour and non-malignant samples is usually performed to iden-
and/or proteomics profiling has provided large and comprehensive tify tumour-specific non-synonymous somatic mutations, followed by
public resources supporting genome-wide screening for CAR targets RNA-seq and mass spectrometry to confirm expression of the mutated
across diverse tissue, cell and cancer types. However, averaging gene or mRNAs and peptides169. Several neoantigens have already been adopted
protein expression data derived from bulk tissues might mask signals as promising CAR targets for the treatment of haematological or solid
from rare cell populations, with potentially important implications for malignancies41,43. For example, EGFR variant III (EGFRvIII) is a neoanti-
the efficacy and toxicity of CAR T cell therapy. gen specifically expressed in certain solid tumours, including a subset
Single-cell technologies, such as scRNA-seq, enable omics profil- of glioblastomas, leading to ongoing clinical trials of EGFRvIII-directed
ing at unprecedented resolution for CAR target discovery, including CAR T cells (such as NCT03726515, NCT03283631 and NCT03638206).
the identification of potential safety signals. For example, an analysis To date, high-throughput multidimensional omics approaches have
of three independent large-scale scRNA-seq datasets derived from enabled the identification of a large number of neoantigens169–172,
human brain tissue samples identified a rare population of mural cells providing a rich source of tumour-specific targets that could be further
expressing CD19, which might contribute to on-target, off-tumour developed for CAR T cell therapy.
neurotoxicity from CD19-directed CAR T cell therapy89. Similarly, an
analysis of two extensive scRNA-seq datasets to predict the on-target, Identification of combinatorial CAR targets
off-tumour toxicity landscape of 591 different CAR targets across a wide Adequately discriminating between tumour and non-malignant cells
range of tissue and cell types identified many as ‘potentially risky genes’ using a single CAR target has proved challenging owing not only
(including EGFR)90; a data portal, CARTSC, was also made available as a to on-target, off-tumour toxicities but also to antigen escape that
resource providing data on CAR target expression at the single-cell level. results in therapy resistance38,100,104,173. This issue can potentially be

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 217


Review article

overcome by combining multiple CAR targets using Boolean “AND” straightforward, requiring integration with data on the human sur-
and/or “AND-NOT” logic gating to increase tumour-targeting speci- faceome190 and validation of target expression through proteomics
ficity and thus reduce toxicity174–178 (Fig. 2). In brief, CAR T cells with techniques (such as flow cytometry). Furthermore, expression of the
“AND” logic gates can be triggered only if both target antigens are simul- target protein on the cell surface does not necessarily correlate with
taneously expressed on the tumour cells, whereas CAR T cells with clinical responses, for example, owing to highly heterogeneous antigen
“AND-NOT” logic gates can be activated only in the presence of a target expression in tumour cells191, warranting careful evaluation of CAR
antigen expressed on tumour cells and the absence of an antigen nor- T cell products with novel targets in rigorous preclinical studies and
mally expressed on non-malignant cells. Multi-omics data integration early phase clinical trials.
has demonstrated the potential to improve CAR T cell design through
the identification of combinatorial CAR targets based on expression Understanding and enhancing CAR T cell efficacy
profiles38,100,104,173. For example, an integrated analysis of extensive tran- and persistence
scriptomics and proteomics datasets derived from non-malignant and A comprehensive understanding of the biological factors that affect
malignant tissues was performed to propose optimal combinatorial the antitumour efficacy of CAR T cells is crucial to the development of
CAR targets in acute myeloid leukaemia, with four possible “AND” pairs strategies to achieve robust and durable therapeutic responses and
fulfilling the stringent efficacy and safety criteria104. A comparative maximize the clinical benefit. The feasibility of integrating bulk and/or
transcriptomics analysis of 3,567 genes encoding surface proteins, single-cell multidimensional omics data to study the key features
based on mRNA expression data derived from tumours included in The related to treatment efficacy has been clearly demonstrated. The main
Cancer Genome Atlas (TCGA) and non-malignant tissues included in determinants of the efficacy and persistence of CAR T cells can be clas-
GTEx, identified 179 “AND” and 443 “AND-NOT” pairs of CAR targets100. sified into four broad categories: T cell states and phenotypes, tumour
Other studies have similarly applied multi-omics data to discover dual cell characteristics, the TME and the microbiota (Fig. 3 and Table 2).
targets for “AND” or “AND-NOT” logic-gate CARs38,173. This strategy can
also be applied to identify more complicated combinations of CAR tar- T cell states and phenotypes
gets such as triple targets. For example, large-scale mRNA expression Pre-manufacture T cell features. The heterogeneity of source T cells
data on 33 tumour types from TCGA and 34 non-malignant tissues from has a crucial role in determining the subsequent in vivo persistence of
GTEx have been leveraged in a comprehensive computational screen of the autologous CAR product28,30,192 (Fig. 3a). For example, an integrative
2,358 predicted surface genes to investigate potential combinations bulk and single-cell profiling study of pre-manufacture T cell popula-
of two or three target antigens for “AND” or “AND-NOT” logic-gated tions revealed that a higher proportion of naive and early memory
CARs173. This study demonstrated the better discriminatory power of T cells, especially in the CD8+ compartment, is associated with longer
combinatorial CAR targets compared with single targets173; data on CAR T cell persistence and greater efficacy in patients with B cell malig-
the performance of all predicted antigen pairs are available through the nancies28. Moreover, TCF7 (TCF1) network activity in effector T cells
Antigen Explorer portal. Although Boolean logic-gated CARs have not was found to correlate with long-term CAR T cell persistence, whereas
yet been used within the clinical setting, other combinatorial antigen- upregulation of genes associated with the type I interferon response,
targeting strategies that mitigate the potential for antigen escape, such as RSAD2, IRF7, MX1, ISG15, OASL and IFIT3, was associated with
for example, those predicated on the expression of CLL1 and CD33 in poor CAR T cell persistence.
acute myeloid leukaemia179,180, CD19 and CD22 in B-ALL and LBCL181,
and BCMA and GPRC5D in multiple myeloma182, have shown promise in CAR T cell features. The composition of the CAR T cell products
enhancing the antitumour activity of CAR T cells in preclinical and/or infused into patients is also highly heterogeneous, and specific CAR
clinical studies. T cell populations have been associated with favourable or unfavour-
Single-cell expression data also provide high resolution for pre- able responses15,30,97,117,193,194 (Fig. 3a). For example, bulk transcriptomics
dicting the efficacy and safety of CAR T cells with combinatorial targets. profiling of CAR T cells before infusion into 41 patients with advanced-
For example, an analysis90 using scRNA-seq data has predicted potential stage chronic lymphocytic leukaemia (CLL) revealed that patients with
toxicity risk for a predefined list of CAR target pairs identified based on a subsequent complete response (CR) had CAR T cells enriched for early
bulk expression data38. Theoretically, full utilization of comprehensive memory programmes and expression of IL-6–STAT3 pathway genes30.
single-cell multi-omics data from both tumour and non-malignant tis- By contrast, CAR T cells of patients who had only a partial response or
sues would be required to nominate optimal target antigens for CAR of non-responders were enriched for the expression of genes associ-
T cell therapies and thus limit the risk of toxicities. Several scRNA-seq ated with T cell differentiation and exhaustion, aerobic glycolysis and
data resources provide a human non-malignant cell atlas encompass- apoptosis30. Features of the CD8+ CAR T cell subset are crucial deter-
ing multiple organs and cell types183–188, and large-scale, pan-cancer, minants of therapeutic efficacy, especially at the initial stage of the
single-cell atlases are in development such as those being mapped by antitumour response. In a single-cell multidimensional analysis of CAR
the Human Tumour Atlas Network189. However, single-cell technolo- T cell infusion products received by patients with relapsed and/or
gies also have limitations, including the large fraction of zero values refractory LBCL, enrichment for memory CD8+ CAR T cells was associ-
of scRNA-seq measurements, which could reflect either transcript ated with a CR at 3 months, whereas CAR T cell products enriched for
drop-out or a true absence of gene expression. At present, the discovery exhausted CD8+ T cells and with low levels of TCR clonotypic diversity
of optimal CAR T cell targets with both high efficacy and safety might were associated with a partial response or progressive disease15.
best and most feasibly be accomplished by first nominating promis- The composition of CAR T cells also affects therapeutic persistence
ing targets that are highly and ubiquitously expressed in tumour cells and/or long-term disease remission. In a study involving 12 patients with
based on bulk omics data, then using single-cell omics data to predict B-ALL, combined scRNA-seq and cellular indexing of transcriptomes
the risk of on-target, off-tumour toxicities for the nominated targets. and epitopes by sequencing (CITE-seq) of the CD19-directed CAR T cell
Notably, identification of CAR targets based on RNA-seq data is not infusion products indicated that deficiency of T helper 2 cell function

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 218


Review article

a T cell states and phenotypes b Tumour cell characteristics


Pre-manufacture T cells
Antigen dependent
Memory Naive
Gene
Heterogeneity expresssion
Target
TF gene Gene Gene Gene
mutation splicing
Promoter CD19

Composition (for
example, memory or
exhaustion states)
Epitope masking

Expression of Antigen loss


Gene CAR T cells
specific genes Gene

Epigenetic Antigen independent


signature
Mutations and/or
Fas copy-number variations
Metabolic
↑ OXPHOS
state
FADD FADD

CAR integration CAR Caspase 8 Caspase 8


site Genomic alteration
(for example, of
Caspase 8
TP53 or FAS)
Different CAR e Proteome Active
intracellular m
lo Epigenome Cell death/apoptosis-
signalling
bo

related pathway
Genome
ta

(for example, Fas expression)


Me

ire
Different rto
CAR T cell pe ns cript
om
ra e
re

products
TCR

c Tumour microenvironment d Microbiota

Increase CAR T cell infiltration


e
m
e
lo
m
bo

a
io

et
ob

M r
ic
M

Transduction of Inhibition of
specific gene specific protein Diversity and composition

Gut
Strengthen CAR T cell against immunosuppression microbiome
Myeloid
Treg cell
cell Butyrate Pentanoate

+
Immune-
checkpoint Combination Co-engineering Microbial metabolites
inhibitors Tumour microbiome

Fig. 3 | Multidimensional omics characterization of molecular features associated data can reveal crucial features of the hostile tumour microenvironment that can
with the efficacy of CAR T cell therapy. a, T cell states and phenotypes, including be leveraged to enhance CAR T cell efficacy, for example, through interventions to
various features of both pre-manufacture T cells and chimeric antigen receptor (CAR) increase CAR T cell infiltration and/or resistance to immunosuppression. d, Micro­
T cell products, can be evaluated across multiple dimensions using omics technologies. biomics and metabolomics data can be used to identify potential associations between
b, Multidimensional omics data can also provide insights on tumour cell characteristics CAR T cell efficacy and the gut microbiota and/or microbial metabolites. FADD,
associated with response or resistance to CAR T cell therapy, classified into antigen- Fas-as­so­ciated death domain protein; OXPHOS, oxidative phosphorylation;
dependent and antigen-independent factors. c, Transcriptomics and proteomics TCR, T cell receptor; TF, transcription factor; Treg, regulatory T.

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 219


Review article

Table 2 | Examples of cellular features associated with CAR T cell efficacy identified using multi-omics data

Characteristic CAR T cell Cancer types Data types Key findings Ref.
targets

Pre-manufacture T cells
Heterogeneity CD19 B-ALL and Hodgkin RNA-seq, scRNA- A higher proportion of naive and early memory T cells was 28

lymphoma in patients seq, CITE-seq and correlated with longer CAR T cell persistence and greater
scATAC-seq efficacy
CAR T cells
Composition CD19 LBCL in patients scRNA-seq, scTCR-seq Memory signature of CD8+ T cells in the infusion product was 15

and DNA-seq enriched in patients with a complete response; patients with a


partial response or progressive disease had CD8+ T cells enriched
for an exhaustion signature and with low TCR clonotypic diversity
CD19 CLL in patients Mass cytometry, CITE- CD8+ CAR T cells were prominent at the initial time point of 97

seq, TCR-seq, scRNA- therapeutic response; cytotoxic characteristics of CD4+ CAR


seq and scTCR-seq T cells were key determinants of long-term remission and
CAR T cell persistence
Expression of CD19 Melanoma, thymoma and ATAC-seq, RNA-seq and Knocking out the three NR4A family transcription factors in CAR 29

specific genes colon adenocarcinoma in scRNA-seq T cells enhanced their antitumour activity
mouse models
Epigenetic CD19 B-ALL and NHL in DNA methylation A specific DNA methylation signature (EPICART) was associated 33

signature patients microarray with favourable clinical outcomes


Metabolic state CD22, BCMA Leukaemia, myeloma and CRISPR gain-of-function CAR T cells overexpressing PRODH2 had enhanced metabolic 34

or HER2 breast cancer in mouse screen, RNA-seq and and immune function as well as antitumour activity
models mass cytometry
CAR integration CD19 CLL in patients RNA-seq, DNA-seq and Integration of CAR sequence into the genome directly disrupted 62

site ATAC-seq the expression of TET2, which led to an enhanced antitumour


response
Intracellular CD19 or B cell lymphoma and Mass spectrometry T cells expressing a CD28–CD3ζ CAR including an additional 109

signalling mesothelin pancreatic cancer in CD3ε signalling module had reduced cytokine release and
domain mouse models enhanced antitumour activity and persistence
Tumour cells
Antigen- CD19 B-ALL in patients DNA-seq and RNA-seq CD19 mutations or mis-splicing result in loss of the CAR target 81

dependent antigen from the cell surface and disease relapse


resistance
CD19 B-ALL in patients RNA-seq and ChIP-seq Lineage switching of B-ALL to a CD19-negative myeloid 205

phenotype resulted in disease relapse


Antigen- CD19 B-ALL in patients Genome-wide loss- Knocking out genes involved in pro-apoptotic death receptor 55

independent of-function screen, signalling in tumour cells reduced their susceptibility to CAR
resistance ATAC-seq, RNA-seq and T cell-mediated cytotoxicity
scRNA-seq
CD19 LBCL in patients DNA-seq and RNA-seq TP53 alterations (mutations and/or copy-number loss) in tumour 44

cells were associated with poor complete response rates and


unfavourable overall survival
Tumour microenvironment
To increase CAR EGFRvIII Glioblastoma in mouse RNA-seq Inhibition of PAK4 improved T cell infiltration and sensitized 35

T cell infiltration models glioblastomas to CAR T cell therapy


To strengthen CD19 and B-ALL and ovarian cancer Mass cytometry IL-18-secreting CAR T cells could activate endogenous immune 222

CAR T cells MUC16 in mouse models cells and modulate the tumour microenvironment, thereby
against immune promoting the antitumour immune response
suppression
Microbiota
Diversity and CD19 B-ALL and NHL in 16S rRNA-seq and Baseline α-diversity was lower in CAR T cell recipients than in 141

composition patients metagenomics shotgun individuals without cancer; the abundance of specific bacterial
sequencing taxa of Lachnospiraceae, Ruminococcaceae and Bacteroidaceae
was higher in patients who had a complete response
Microbial ROR1 Pancreatic cancer in Mass spectrometry The microbial metabolites pentanoate and butyrate could 223

metabolites mouse models enhance the antitumour activity of CAR T cells


16S rRNA-seq, 16S ribosomal RNA sequencing; ATAC-seq, assay for transposase-accessible chromatin with high-throughput sequencing; B-ALL, B cell acute lymphocytic leukaemia;
CAR, chimeric antigen receptor; ChIP-sequencing, chromatin immunoprecipitation sequencing; CITE-seq, cellular indexing of transcriptomes and epitopes by sequencing; CLL, chronic
lymphocytic leukaemia; DNA-seq, DNA sequencing; EGFRvIII, EGFR variant III; LBCL, large B cell lymphoma; NHL, non-Hodgkin lymphoma; RNA-seq, RNA sequencing; scATAC-seq, single-cell
ATAC-seq; scRNA-seq, single-cell RNA-seq; scTCR-seq, single-cell T cell receptor sequencing; TCR, T cell receptor; TCR-seq, TCR sequencing.

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 220


Review article

and enrichment for effector-related phenotypes is predictive of CD19+ The intracellular signalling domain of the CAR construct can also
disease relapse194. Furthermore, data suggest that certain CD4+ CAR influence CAR T cell activity (Fig. 3a). In a simultaneous examination
T cell states are a predominant requirement for long-term remission of the dynamics of immunoreceptor tyrosine-based activation motif
following CD19-directed CAR T cell therapy. In a longitudinal study, (ITAM) phosphorylation within the CD3ε, CD3δ, CD3γ and CD3ζ signal-
both bulk and single-cell multidimensional omics data were used to ling subunits of the TCR using quantitative mass spectrometry, high
characterize the functional and molecular features of CD19-directed levels of mono-phosphorylation and a basic residue-rich sequence
CAR T cells in two patients with CLL who had durable CRs lasting more of CD3ε were found to restrain TCR signalling109. Notably, addition of
than a decade; persistent CD4+ CAR T cell clones with cytotoxic, prolif- the CD3ε-derived ITAM–basic residue-rich sequence region into a
erating and metabolically active phenotypes were identified as a key CD19–CD28−CD3ζ CAR construct resulted in a CAR T cell product asso-
determinant of long-term CAR T persistence97. ciated with reduced levels of cytokine release, enhanced antitumour
CAR T cell-intrinsic features, including expression of specific responses and increased persistence in a mouse xenograft model of
genes29,32,195,196 (such as key transcription factors), epigenetic signa- B cell lymphoma. Indeed, different CAR T products have different
tures33 and metabolic states34, are associated with therapeutic efficacy phenotypes and expansion characteristics. Axicabtagene ciloleucel
(Fig. 3a). For example, single-cell transcriptomics and chromatin acces- (axi-cel) and tisagenlecleucel, for example, differ not only in the source
sibility profiling data implicate the transcription factors NR4A1, NR4A2 of their intracellular co-stimulatory domains (CD28 versus 4-1BB) but
and NR4A3 in CAR T cell dysfunction29. Moreover, ATAC-seq of Nr4a also in their hinge–transmembrane domains (CD28 versus CD8α)
triple-knockout CAR T cells showed enrichment of accessible chro- and their integration vectors (gammaretrovirus versus lentivirus)199.
matin regions containing binding motifs for the transcription factors In a scRNA-seq analysis of the infusion products and 105 peripheral
NF-κB and AP1 (ref. 29). Data from a multidimensional transcriptomics, blood mononuclear cell samples collected at different pre-treatment
epigenomics and single-cell proteomics study indicate that the tran- and post-treatment time points from 19 patients receiving axi-cel and
scription factor BATF can prevent CAR T cell exhaustion and facilitates 13 receiving tisagenlecleucel, expansion of central memory CD8+ cells
the transition of phenotypes and transcriptional profiles towards an was only associated with responses to tisagenlecleucel; responders to
effector-like state196. In an alternative approach to gene expression axi-cel had more heterogeneous T cell populations, while enrichment
analysis, a predictive DNA methylation signature termed EPICART has of CAR-expressing regulatory T (Treg) cells in the axi-cel infusion prod-
been generated using DNA methylation array data from 114 patients uct were associated with a lack of response24. Together, these findings
with B cell malignancies treated with CD19-directed CAR T cells33. underscore the application of multi-omics data in characterizing the
Pre-infusion CAR T cells positive for this signature were associated with differences between distinct CAR designs and cell products.
better clinical outcomes in both the initial discovery cohort (n = 77)
and an independent validation cohort (n = 35) and were enriched for Tumour cell characteristics
naive-like or early memory T cell phenotypes33. Antigen-dependent resistance. Tumour cell characteristics, includ-
The metabolic state of the CAR T cells might influence antitumour ing both antigen-dependent and antigen-independent mechanisms,
activity and toxicity (Fig. 3a). A guide RNA-based CRISPR gain-of- are crucial drivers of resistance to CAR T cells166,200,201 (Fig. 3b). Antigen
function screening platform has been developed and applied to primary escape in tumours (that is, loss of the target epitope) is the leading
CD8+ T cells, resulting in the identification of upregulation of PRODH2, cause of antigen-dependent resistance to CAR T cells and has been
which encodes hydroxyproline dehydrogenase (an enzyme involved shown to occur via downregulation202, mutation160, alternative splic-
in proline metabolism), as a means to augment CAR T cell function34. ing81,203,204, lineage switching205,206 or antigen masking207. A DNA-seq
Transcriptomics and metabolomics profiling of PRODH2-knock-in and RNA-seq analysis found that all 12 evaluated patients relapsing
CAR T cells revealed enhanced metabolic and immunological activity, with CD19-negative B-ALL after CD19-directed CAR T cell therapy had
and these cells had improved antitumour activity in multiple mouse somatic mutations in CD19 exons 2–5, which are postulated to result
models34. Furthermore, CAR T cell metabolic activity might lead to in truncation of CD19 and subsequent loss of expression on the cell
imbalances, such as hypophosphataemia, that contribute to ICANS197. surface160. An RNA-seq study demonstrated alternative splicing of
Integration of the CAR transgene into the genome can affect CD19 with exon 2 skipping, leading to a loss of the extracellular CD19
key genes, thereby influencing the clinical outcome of CAR T cell epitope81. By contrast, scRNA-seq in a patient with relapsing B-ALL
therapy62,198 (Fig. 3a). For example, insertion of a CAR sequence into revealed antigen loss of CD19 via mis-splicing that resulted in a non-
T cells from a patient with CLL disrupted the transcription of TET2 functional CD19 transcript retaining intron 2, and this subclone was
(encoding a methylcytosine dioxygenase involved in epigenetic regu- present before CAR T cell treatment203. A study using flow cytometry
lation) and resulted in an enhanced antitumour response62. Further reported switching of B-ALL to a CD19– myeloid lineage in four patients
transcriptomics and epigenomics analyses revealed that the patient after CD19-directed CAR T cell therapy205. Combined RNA-seq and
had a pre-existing hypomorphic mutation in the second TET2 allele and ChIP-seq data from mouse models provided further validation that
that TET2-disrupted CAR T cells had increased expression of effector the myeloid lineage switch was triggered following CD19-targeted CAR
molecules, such as perforin and granzyme B, as well as a central mem- T cell infusion205. In addition, combined genomics and transcriptomics
ory state that prevented cells from becoming terminally exhausted. sequencing has uncovered a novel but rare mechanism of epitope
In a high-throughput sequencing study of CAR vector-integration masking related to resistance to CAR T cell therapy: the CAR gene was
sites in 40 patients with CLL or B-ALL, integration sites in patients transduced into a single leukaemic B cell by accident, and expression
who responded to therapy were enriched in genes related to pathways of the CD19-directed CAR on the progeny of this leukaemic clone pre-
mediating T cell proliferation, such as phosphatidylinositol, cyclic vented recognition by, and thus conferred resistance to, CAR T cells
AMP or TCR signalling, or covalent chromatin modification, suggest- via competitive binding to CD19 on leukaemic cells in cis207. These
ing that insertional mutagenesis in these genes might lead to better results demonstrate the complexity of antigen escape mechanisms in
clinical outcomes198. B-ALL. However, despite approximately 28% of LBCLs having reduced

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 221


Review article

CD19 expression at relapse after CD19-directed CAR T cell therapy202, findings suggest that intratumoural myeloid cells and cytokine levels
mutation and alternative splicing are not observed and the mechanisms are associated with response to CD19-directed CAR T cells. Here, we
remain to be defined; thus, different mechanisms of antigen escape are focus on the application of multidimensional omics data to obtain an
likely to exist in distinct malignancies. The application of genomics, in-depth understanding of the hostile TME in order to facilitate the
transcriptomics and/or proteomics profiling will be important to elu- development of new approaches to enhance the intratumoural abun-
cidate the mechanisms of antigen escape across diverse contexts and dance, persistence and activity of CAR T cells and thereby improve
to develop strategies to circumvent antigen-dependent resistance, for their clinical efficacy (Fig. 3c).
example, through the design of dual-targeted CAR T cell products181,182.
Increasing CAR T cell infiltration. Omics data have been utilized to
Antigen-independent resistance. Antigen-independent resistance identify key regulators that could be leveraged to increase CAR T cell
resulting from the intrinsic insensitivity of tumour cells to the mecha- infiltration into the TME. In a mouse model of breast cancer, the stimu-
nisms of CAR T cell-mediated cytotoxicity also contributes substan- lator of interferon genes (STING) agonist DMXAA greatly enhanced
tially to treatment failure (Fig. 3b). Emerging evidence indicates that CAR T cell recruitment to and persistence in the TME, with scRNA-seq
dysfunctional death receptor signalling in tumour cells can lead to revealing favourable shifts in the chemokine milieu and the balance
resistance to CAR T cell cytotoxicity. In a genome-wide CRISPR–Cas9 of immunostimulatory versus immunosuppressive myeloid cells214.
knockout screen, disruption of genes involved in pro-apoptotic death In a kinome-wide screening study, RNA-seq revealed that PAK4 has an
receptor signalling (including FADD, BID, CASP8 and TNFRSF10B) in important role in regulating mesenchymal-like transcription activa-
B-ALL cell lines conferred resistance to CD19-directed CAR T cells, tion in glioblastoma-derived endothelial cells; inhibition of this kinase
which was further reinforced by induction of CAR T cell dysfunction as a improved T cell infiltration and sensitized glioblastoma tumours to
result of antigen persistence55. Conversely, knocking out anti-apoptotic EGFRvIII-directed CAR T cells in a mouse model35.
genes (such as CFLAR, TRAF2 and BIRC2) in leukaemic cells increased
their susceptibility to CAR T cell-mediated cytotoxicity55. Similarly, Strengthening CAR T cells against immunosuppression. Multi-
another CRISPR–Cas9 screen demonstrated that expression of the omics analyses also present a powerful approach for identifying key
death receptor Fas on tumour cells is essential for antigen-dependent immunosuppressive factors in the TME, thereby enabling the engi-
CAR T cell-mediated killing as well as for bystander T cell-mediated neering of CAR T cells capable of resisting such factors1. A single-cell
killing of antigen-negative tumour cells208. Notably, DNA-seq and RNA- transcriptomics analysis of oesophageal squamous cell carcinomas
seq data indicate that patients with LBCL harbouring FAS deletions45 or indicated that PD-L1 is upregulated on tolerogenic dendritic cells in the
with low FAS expression208 have inferior survival outcomes following TME215, and data from a pan-cancer scRNA-seq atlas of T cells suggested
CD19-directed CAR T cell therapy. that binding of PD-L1 to PD-1 (ref. 216) inhibits T cell cytotoxicity and
Other genomics alterations within tumour cells have also been facilitates immune evasion, underscoring the known role of the PD-L1–
correlated with poor outcomes following CAR T therapy. Integrated PD-1 axis as a key mediator of immunosuppression in the TME. There-
targeted DNA-seq of pre-treatment tumour samples from 82 patients fore, the efficacy of CAR T cells against poorly responding tumours
with relapsed and/or refractory LBCL receiving CD19-directed CAR might be enhanced by combining CAR T cells with anti-PD-(L)1 anti-
T cell therapy demonstrated that TP53 aberrations (mutations and/or bodies217, engineering PD-1-blocking CAR T cells218 or CAR T cells that
copy-number losses) are associated with reduced overall survival but express PD-1 dominant-negative receptors219, or genetically disrupting
not with progression-free survival44. Bulk transcriptomics data from PD-1 expression in CAR T cells220.
a separate cohort of 562 patients further suggested that TP53 altera- A pan-cancer analysis based on RNA-seq data from TCGA and GTEx
tions in tumours impair processes such as interferon signalling, death revealed that FASLG (encoding Fas ligand; FasL) is often overexpressed
receptor signalling and CD8+ T cell infiltration, which are important in the TME221. Accordingly, CAR T cells co-engineered to express dom-
for CAR T cell-mediated cytotoxicity44. TP53 alterations are also linked inant-negative variants of Fas (the receptor of FasL that is typically
with increased genomics complexity in a variety of contexts, including highly expressed on autologous T cells used for adoptive cell therapy)
LBCL209, and high levels of genomics complexity have been associated had increased persistence and antitumour efficacy in mouse models of
with poor outcomes in patients with LBCL receiving CD19-directed CAR various solid and haematological malignancies221. In addition, a CyTOF
T cells45,46. Taken together, these studies emphasize that integration of analysis demonstrated that IL-18-secreting CAR T cells can modulate
multidimensional omics data is crucial to reveal the tumour-intrinsic the TME and enhance the activity of endogenous immune cells (for
mechanisms of resistance to CAR T cell therapy. example, promoting endogenous CD8+ T cells with a central memory
phenotype, macrophages with an M1 phenotype, and dendritic cells
The TME with a more mature and activated phenotype) in diverse mouse models,
The TME typically contains diverse immunosuppressive cells, including thereby promoting antitumour immune responses222. Taken together,
Treg cells, cancer-associated fibroblasts, tumour-associated macro­ these studies demonstrate that leveraging multidimensional omics
phages and myeloid-derived suppressor cells, and cytokines (such data can greatly help to overcome factors in the TME that limit the
as TGFβ, IL-10, IL-4 and VEGF) that might limit CAR T cell infiltration, effectiveness of CAR T cell therapy for solid tumours.
proliferation and effector function, and ultimately lead to CAR T cell
exhaustion16,210–212. The challenges to CAR T cell therapy posed by The microbiota
the immunosuppressive TME and the engineering strategies aiming The microbiome is emerging as a crucial factor affecting the efficacy
to overcome them have been extensively reviewed elsewhere166,213. of CAR T cell therapy (Fig. 3d). In a multicentre study involving a pro-
Interrogation of the TME in patients treated with CAR T cells has been spective cohort of 48 patients with B cell lymphomas or leukaemias
limited to targeted gene expression assays25,210 and has not yet receiving CD19-directed CAR T cell therapy, 16S rRNA-seq and metagen-
been comprehensively explored with omics approaches, although the omics shotgun sequencing of the baseline faecal samples revealed a

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 222


Review article

considerably lower level of α-diversity and a substantially different com- demonstrated the potential of multi-omics profiling to elucidate the
position of the gut microbiota compared with that of individuals with- mechanisms underlying these toxicities and thus inform potential
out cancer141. Furthermore, specific bacterial taxa of Lachnospiraceae, prophylactic and therapeutic strategies.
Ruminococcaceae and Bacteroidaceae were in greater abundance in
patients who had a CR at day 100 than those who did not. Preclinical CRS
data from mouse models of melanoma and pancreatic cancer indi- CRS results from robust cytokine production by CAR T cells follow-
cate that two microbial metabolites identified using metabolomics ing engagement with target cells as well as by subsequently activated
data from the human commensal bacteria Megasphaera massiliensis, endogenous immune cells, which creates a loop of immune activation
namely pentanoate and butyrate, can increase the antitumour activity that can cause a ‘cytokine storm’226,228. Characterization of cytokine lev-
of CAR T cells through metabolic and epigenetic reprogramming223. els and immune cell interactions in the context of CRS has been crucial
Prior knowledge that not only the intestinal microbiota but also the to understanding its aetiology (Fig. 4a). Transcriptomics approaches
intratumour microbiota can have multiple effects on human cancers have been used to determine the key immune cell types and/or cellular
along with the development of technologies that enable quantitative interactions that mediate this excessive cytokine release. In a mouse
analyses of the microbiome132,142 will facilitate further investigations model of CAR T cell-induced CRS, bulk RNA-seq of myeloid cells iso-
to characterize the influence of the microbiota on the efficacy of CAR lated by fluorescence-activated cell sorting revealed that increased
T cell therapy. expression of IL-1 receptors and IL-6 by these cells, and particularly by
macrophages, is associated with greater severity of CRS37. Moreover,
Minimizing CAR T cell-related toxicities a scRNA-seq analysis of CD45+ leukocytes from a humanized mouse
The toxicities of CAR T cell therapies, which can be serious or even model of high-burden leukaemia implicated circulating human mono-
fatal18, remain a substantial challenge to widespread clinical applica- cytes as the primary source of IL-6 and IL-1 released following CAR
tion224–226. In the process of killing antigen-expressing target cells, CAR T cell infusion, with a time-course analysis indicating that production
T cells induce a series of inflammatory responses via the secretion of IL-1 preceded IL-6 during the development of CRS36. Furthermore, a
of various cytokines and chemokines — a double-edged sword that cytokine signature generated through proteomics profiling of serum
amplifies antitumour immune responses but can also cause severe samples from 51 patients receiving CD19-directed CAR T cell therapy
toxicities16. The most common CAR T cell-induced adverse events accurately predicted the likelihood of severe CRS, potentially enabling
include CRS, ICANS and prolonged cytopenias227. Several studies have early intervention229.

b ICANS

Blood vessel Mural cell

CAR
T cell Microbiome Monocyte-like
composition CAR T cells Density of Treg cells

a CRS c On-target, off-tumour


toxicities
CAR T cell Tumour cell

Non-malignant
cell

Microbiome
IL-1, Cytokines
IL-6
nscriptome
Tra
Pr
e
om

ote

CAR T cell
Prote

ome

Monocytes/ Tumour
myeloid cells cell
CAR T cell
therapy-
related
toxicities

Fig. 4 | Summary of factors related to major categories of CAR T cell-induced neurotoxicity syndrome (ICANS) have been identified using transcriptomics and
toxicities revealed by multi-omics data. a, Factors involved in chimeric microbiomics data. c, On-target, off-tumour effects of CAR T cell products can
antigen receptor (CAR) T cell-associated cytokine-release syndrome (CRS) that also be characterized, and potentially predicted, through transcriptomics and
have been characterized through transcriptomics and proteomics. b, Factors proteomics analyses. Treg, regulatory T.
related to the risk and/or mechanism of immune effector cell-associated

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 223


Review article

ICANS new molecular insights into CAR T cell therapy. Multi-omics profiling
ICANS is another major toxicity of CD19-directed CAR T cell ther- of tumour and non-malignant tissues provides a feasible strategy for
apy19,227,230. Targeted gene expression analysis has identified an asso- large-scale screening of CAR targets and logical designs. Meanwhile,
ciation between a high pre-treatment intratumoural density of Treg cells characterization of T cells, tumour cells, the TME and the microbiota
and reduced risk of ICANS in patients with LBCL25, and targeted single- can decipher the molecular features and pathways that determine
cell cytokine analysis has shown that higher frequencies of polyfunc- therapeutic efficacy as well as the potential underlying mechanisms and
tional IL-17A-producing T cells within infusion products are associated pivotal regulators of different categories of toxicities associated with
with increased risk of high-grade ICANS. Moreover, scRNA-seq of CD19- CAR T cell therapy. The current FDA-approved CAR T cell products are
directed CAR T cell infusion products for patients with LBCL revealed manufactured from autologous T cells owing to the advantage of avoid-
a small subset of cells with a monocyte-like transcriptional signature ing graft versus host disease, yet such products have disadvantages
within the infusion products of patients with high-grade ICANS15. Nota- related to long manufacturing times and impaired functional fitness of
bly, these cells express high levels of IL-1β; this finding, together with patient-derived T cells231. Notably, multi-omics studies of donor-derived
the independent aforementioned support for the pathophysiological allogeneic CAR T cell products and their comparison with autologous
role of IL-1 signalling in CRS36,37 and ICANS15,36, has prompted clinical products using transcriptomics, proteomics and metabolomics data
investigation of an IL-1 receptor antagonist for prophylaxis of CRS and have demonstrated the impaired metabolic fitness of autologous CAR
ICANS (NCT04432506). In addition, scRNA-seq data from human brain T cells as well as a higher level of activation in allogeneic CAR T cells,
tissue suggest that CD19 expression on brain mural cells might also indicating a greater antitumour potential87,130,232. These studies have
be a target for CD19-directed CAR T cells89. Furthermore, 16S rRNA- also advanced the mechanistic understanding of allogeneic products
seq and metagenomics shotgun sequencing of stool samples from and delineated a promising future for ‘off the shelf’ CAR T cells.
45 patients with B cell malignancies receiving CD19-directed CAR T cells Advances in single-cell technologies have greatly facilitated explo-
demonstrated an association between ICANS and faecal microbiota ration of the remarkable diversity of cellular phenotypes and states
composition141. The mechanisms underlying ICANS are multifactorial among both tumour and immune cells in the TME, expanding our col-
and remain poorly understood; however, a clearer understanding might lective knowledge in the field of immuno-oncology93,233. The parallel
be obtained by leveraging multi-omics data (Fig. 4b). improvements in analytical algorithms to interpret single-cell multi-
omics data have supported important advances in single-cell tech-
On-target, off-tumour toxicities nologies70,113,234–237. Further application of single-cell technologies to
The targets of most CAR T cell products currently used in the clinic study various aspects of CAR T cell therapy is likely to provide rich and
and/or being tested in clinical trials are expressed to some extent on high-resolution information, offering clinically relevant insights into
non-malignant cells; therefore, CAR T cells can cause damage to non- treatment strategies to maximize the potential for durable remissions.
malignant tissues and result in on-target, off-tumour toxicities27,38,227, Furthermore, spatial profiling technologies enable in situ cell charac-
especially when targeting solid tumours18. Predicting the risk of on- terization and mapping of spatially preserved tissue sections. Although
target, off-tumour toxicities and monitoring patient safety through not yet utilized to study CAR T cell therapy, spatial transcriptomics
multi-omics profiling during treatment could potentially enable more and proteomics have been used to investigate interactions between
efficient management of such adverse effects38. By combining bulk tumour cells and infiltrating immune cells in the context of immune-
transcriptomics and proteomics data, a targetable landscape encom- checkpoint inhibitor therapy in order to explore the clinical relevance
passing >100 single CAR targets and >100,000 logic-gated target pairs of cell localization238,239. These advances in single-cell and spatial multi-
with minimal predicted toxicity has been reported38. Similarly, the omics technologies have demonstrated promise in dissecting the
study of two independent large-scale scRNA-seq datasets90 defined cellular components and their intercellular connections, providing
the safety landscape of 591 CAR targets and 320,884 logic-gated “AND” opportunities to overcome obstacles to effective therapeutic targeting,
design target pairs at the single-cell level. Notably, by leveraging scRNA- including immune evasion and immunosuppression of CAR T cells.
seq to dissect rare cell populations and/or types, several potentially Leveraging valuable multidimensional resources through multi-
risky targets were identified that might result in toxicities affecting omics data integration is also of great relevance in elucidating the evo-
several important organs, including EGFR, prostate stem cell antigen lution and transitions of cells during therapy. For example, combined
and VEGFR2, which were not identified as a safety risk in bulk expres- RNA-seq, CITE-seq and single-cell ATAC-seq data have been used to
sion analyses90. In summary, multi-omics analyses could substantially track the molecular determinants of long-term CAR T cell persistence28.
broaden molecular insights on toxicities and thus facilitate the devel- Furthermore, the integration of omics data from multiple studies
opment of approaches to mitigate the adverse effects of CAR T cell enables a substantially larger sample size and might thus lead to novel
therapies (Fig. 4c). discoveries of important biological processes or features formerly
underappreciated in studies with relatively small sample sizes240. To this
Conclusions end, further optimization of computational frameworks for omics
CAR T cell therapy has transformed the management of B cell malig- data integration is required to fully leverage multi-omics data in future
nancies, yet substantial numbers of patients do not have a clinical research in the realm of CAR T cell therapy and beyond164,241.
response or have disease relapse owing to poor CAR T cell function
and/or resistance of tumour cells to such therapies. Moreover, the Published online: 31 January 2023
applicability of CAR T cell therapies remains limited owing to a series of
challenges that include a lack of appropriate CAR targets, the potential References
for disease relapse and treatment-associated adverse events. Solu- 1. Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current
roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).
tions to these challenges are under exploration and development. 2. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia.
The expanding multi-omics data repertoire could be utilized to obtain N. Engl. J. Med. 371, 1507–1517 (2014).

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 224


Review article

3. Schuster, S. J. et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. 38. MacKay, M. et al. The therapeutic landscape for cells engineered with chimeric antigen
N. Engl. J. Med. 377, 2545–2554 (2017). receptors. Nat. Biotechnol. 38, 233–244 (2020).
4. Wang, M. et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. 39. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science
N. Engl. J. Med. 382, 1331–1342 (2020). 348, 69–74 (2015).
5. Fowler, N. H. et al. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: 40. Yamamoto, T. N., Kishton, R. J. & Restifo, N. P. Developing neoantigen-targeted T cell-
the phase 2 ELARA trial. Nat. Med. 28, 325–332 (2022). based treatments for solid tumors. Nat. Med. 25, 1488–1499 (2019).
6. Berdeja, J. G. et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed 41. Hwang, M. S. et al. Structural engineering of chimeric antigen receptors targeting
chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple HLA-restricted neoantigens. Nat. Commun. 12, 5271 (2021).
myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet 398, 314–324 (2021). 42. Choe, J. H. et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity,
7. Brown, C. E. et al. Regression of glioblastoma after chimeric antigen receptor T-cell and persistence in treating glioblastoma. Sci. Transl. Med. 13, eabe7378 (2021).
therapy. N. Engl. J. Med. 375, 2561–2569 (2016). 43. O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR
8. Beatty, G. L. et al. Activity of mesothelin-specific chimeric antigen receptor T cells against T cells mediates antigen loss and induces adaptive resistance in patients with recurrent
pancreatic carcinoma metastases in a phase 1 trial. Gastroenterology 155, 29–32 (2018). glioblastoma. Sci. Transl. Med. 9, eaaa0984 (2017).
9. Ahmed, N. et al. Human epidermal growth factor receptor 2 (HER2)-specific chimeric 44. Shouval, R. et al. Impact of TP53 genomic alterations in large B-cell lymphoma treated
antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. with CD19-chimeric antigen receptor T-cell therapy. J. Clin. Oncol. 40, 369–381 (2022).
J. Clin. Oncol. 33, 1688–1696 (2015). 45. Cherng, H.-J. J. et al. Risk assessment with low-pass whole-genome sequencing of cell-
10. Qi, C. et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim free DNA before CD19 CAR T-cell therapy for large B-cell lymphoma. Blood 140, 504–515
results. Nat. Med. 28, 1189–1198 (2022). (2022).
11. Narayan, V. et al. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic 46. Jain, M. D. et al. Whole-genome sequencing reveals complex genomic features underlying
castration-resistant prostate cancer: a phase 1 trial. Nat. Med. 28, 724–734 (2022). anti-CD19 CAR T-cell treatment failures in lymphoma. Blood 140, 491–503 (2022).
12. Irving, M., Zoete, V., Bassani-Sternberg, M. & Coukos, G. A roadmap for driving CAR 47. Sworder, B. J. et al. Determinants of resistance to engineered T cell therapies targeting
T cells toward the oncogenic immunopeptidome. Cancer Cell 40, 20–22 (2022). CD19 in large B cell lymphomas. Cancer Cell https://doi.org/10.1016/j.ccell.2022.12.005
13. Leko, V. & Rosenberg, S. A. Identifying and targeting human tumor antigens for (2022).
T cell-based immunotherapy of solid tumors. Cancer Cell 38, 454–472 (2020). 48. Zhou, Y. et al. High-throughput screening of a CRISPR/Cas9 library for functional
14. Delgoffe, G. M. et al. The role of exhaustion in CAR T cell therapy. Cancer Cell 39, genomics in human cells. Nature 509, 487–491 (2014).
885–888 (2021). 49. Bock, C. et al. High-content CRISPR screening. Nat. Rev. Methods Prim. https://doi.org/
15. Deng, Q. et al. Characteristics of anti-CD19 CAR T cell infusion products associated with 10.1038/s43586-021-00093-4 (2022).
efficacy and toxicity in patients with large B cell lymphomas. Nat. Med. 26, 1878–1887 50. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key
(2020). regulators of immune function. Cell 175, 1958–1971.e15 (2018).
16. Hou, A. J., Chen, L. C. & Chen, Y. Y. Navigating CAR-T cells through the solid-tumour 51. Wei, J. et al. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy.
microenvironment. Nat. Rev. Drug Discov. 20, 531–550 (2021). Nature 576, 471–476 (2019).
17. Milone, M. C. et al. Engineering-enhanced CAR T cells for improved cancer therapy. 52. Wang, D. et al. CRISPR screening of CAR T cells and cancer stem cells reveals critical
Nat. Cancer 2, 780–793 (2021). dependencies for cell-based therapies. Cancer Discov. 11, 1192–1211 (2021).
18. Neelapu, S. S. et al. Chimeric antigen receptor T-cell therapy-assessment and 53. Dufva, O. et al. Integrated drug profiling and CRISPR screening identify essential
management of toxicities. Nat. Rev. Clin. Oncol. 15, 47–62 (2018). pathways for CAR T-cell cytotoxicity. Blood 135, 597–609 (2020).
19. Santomasso, B. D. et al. Management of immune-related adverse events in patients 54. Gurusamy, D. et al. Multi-phenotype CRISPR-Cas9 screen identifies p38 kinase as a target
treated with chimeric antigen receptor T-cell therapy: ASCO guideline. J. Clin. Oncol. 39, for adoptive immunotherapies. Cancer Cell 37, 818–833.e9 (2020).
3978–3992 (2021). 55. Singh, N. et al. Impaired death receptor signaling in leukemia causes antigen-
20. Hasin, Y., Seldin, M. & Lusis, A. Multi-omics approaches to disease. Genome Biol. 18, 83 independent resistance by inducing CAR T-cell dysfunction. Cancer Discov. 10, 552–567
(2017). (2020).
21. Deng, Y., Finck, A. & Fan, R. Single-cell omics analyses enabled by microchip 56. Yan, X. et al. CD58 loss in tumor cells confers functional impairment of CAR T cells.
technologies. Annu. Rev. Biomed. Eng. 21, 365–393 (2019). Blood Adv. 6, 5844–5856 (2022).
22. Cantini, L. et al. Benchmarking joint multi-omics dimensionality reduction approaches 57. Roth, T. L. et al. Pooled knockin targeting for genome engineering of cellular
for the study of cancer. Nat. Commun. 12, 124 (2021). immunotherapies. Cell 181, 728–744.e21 (2020).
23. Jiang, P. et al. Big data in basic and translational cancer research. Nat. Rev. Cancer 58. Zebley, C. C., Gottschalk, S. & Youngblood, B. Rewriting history: epigenetic reprogramming
https://doi.org/10.1038/s41568-022-00502-0 (2022). of CD8+ T cell differentiation to enhance immunotherapy. Trends Immunol. 41, 665–675
24. Haradhvala, N. J. et al. Distinct cellular dynamics associated with response to CAR-T (2020).
therapy for refractory B cell lymphoma. Nat. Med. 28, 1848–1859 (2022). 59. Henning, A. N., Roychoudhuri, R. & Restifo, N. P. Epigenetic control of CD8+ T cell
25. Scholler, N. et al. Tumor immune contexture is a determinant of anti-CD19 CAR T cell differentiation. Nat. Rev. Immunol. 18, 340–356 (2018).
efficacy in large B cell lymphoma. Nat. Med. https://doi.org/10.1038/s41591-022-01916-x 60. Akbari, B. et al. Epigenetic strategies to boost CAR T cell therapy. Mol. Ther. 29,
(2022). 2640–2659 (2021).
26. Good, Z. et al. Post-infusion CAR Treg cells identify patients resistant to CD19-CAR 61. Chan, J. D. et al. Cellular networks controlling T cell persistence in adoptive cell therapy.
therapy. Nat. Med. 28, 1860–1871 (2022). Nat. Rev. Immunol. 21, 769–784 (2021).
27. Young, R. M., Engel, N. W., Uslu, U., Wellhausen, N. & June, C. H. Next-generation CAR 62. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted
T-cell therapies. Cancer Discov. 12, 1625–1633 (2022). T cells. Nature 558, 307–312 (2018).
28. Chen, G. M. et al. Integrative bulk and single-cell profiling of premanufacture T-cell 63. Weber, E. W. et al. Transient rest restores functionality in exhausted CAR-T cells through
populations reveals factors mediating long-term persistence of CAR T-cell therapy. epigenetic remodeling. Science 372, eaba1786 (2021).
Cancer Discov. 11, 2186–2199 (2021). 64. Guo, A. et al. cBAF complex components and MYC cooperate early in CD8+ T cell fate.
29. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature Nature 607, 135–141 (2022).
567, 530–534 (2019). 65. Stricker, S. H., Köferle, A. & Beck, S. From profiles to function in epigenomics. Nat. Rev.
30. Fraietta, J. A. et al. Determinants of response and resistance to CD19 chimeric antigen Genet. 18, 51–66 (2016).
receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 24, 563–571 66. Wang, Y. et al. Low-dose decitabine priming endows CAR T cells with enhanced and
(2018). persistent antitumour potential via epigenetic reprogramming. Nat. Commun. 12, 409
31. Xhangolli, I. et al. Single-cell analysis of CAR-T cell activation reveals a mixed TH1/TH2 (2021).
response independent of differentiation. Genomics Proteomics Bioinformatics 17, 129–139 67. Wei, Y. et al. Targeting CDK7 suppresses super enhancer-linked inflammatory genes and
(2019). alleviates CAR T cell-induced cytokine release syndrome. Mol. Cancer 20, 5 (2021).
32. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 68. Gennert, D. G. et al. Dynamic chromatin regulatory landscape of human CAR T cell
576, 293–300 (2019). exhaustion. Proc. Natl Acad. Sci. USA 118, e2104758118 (2021).
33. Garcia-Prieto, C. A. et al. Epigenetic profiling and response to CD19 chimeric antigen 69. Ding, Z.-C. et al. Persistent STAT5 activation reprograms the epigenetic landscape
receptor T-cell therapy in B-cell malignancies. J. Natl Cancer Inst. 114, 436–445 (2022). in CD4+ T cells to drive polyfunctionality and antitumor immunity. Sci. Immunol. 5,
34. Ye, L. et al. A genome-scale gain-of-function CRISPR screen in CD8 T cells identifies eaba5962 (2020).
proline metabolism as a means to enhance CAR-T therapy. Cell Metab. 34, 595–614.e14 70. Preissl, S., Gaulton, K. J. & Ren, B. Characterizing cis-regulatory elements using
(2022). single-cell epigenomics. Nat. Rev. Genet. https://doi.org/10.1038/s41576-022-00509-1
35. Ma, W. et al. Targeting PAK4 to reprogram the vascular microenvironment and improve (2022).
CAR-T immunotherapy for glioblastoma. Nat. Cancer 2, 83–97 (2021). 71. Jiang, P. et al. Single-cell ATAC-seq maps the comprehensive and dynamic chromatin
36. Norelli, M. et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine- accessibility landscape of CAR-T cell dysfunction. Leukemia https://doi.org/10.1038/
release syndrome and neurotoxicity due to CAR T cells. Nat. Med. 24, 739–748 (2018). s41375-022-01676-0 (2022).
37. Giavridis, T. et al. CAR T cell-induced cytokine release syndrome is mediated by 72. Bagaev, A. et al. Conserved pan-cancer microenvironment subtypes predict response
macrophages and abated by IL-1 blockade. Nat. Med. 24, 731–738 (2018). to immunotherapy. Cancer Cell 39, 845–865.e7 (2021).

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 225


Review article

73. Hong, M. et al. RNA sequencing: new technologies and applications in cancer research. 109. Wu, W. et al. Multiple signaling roles of CD3ε and its application in CAR-T cell therapy.
J. Hematol. Oncol. 13, 166 (2020). Cell 182, 855–871.e23 (2020).
74. Xiang, Y., Ye, Y., Zhang, Z. & Han, L. Maximizing the utility of cancer transcriptomic data. 110. Lombard-Banek, C. & Schiel, J. E. Mass spectrometry advances and perspectives for the
Trends Cancer 4, 823–837 (2018). characterization of emerging adoptive cell therapies. Molecules 25, 1396 (2020).
75. Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription 111. Saeys, Y., Van Gassen, S. & Lambrecht, B. N. Computational flow cytometry: helping
factors to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410–12415 to make sense of high-dimensional immunology data. Nat. Rev. Immunol. 16, 449–462
(2019). (2016).
76. Good, C. R. et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 112. Bonilla, D. L., Reinin, G. & Chua, E. Full spectrum flow cytometry as a powerful
6081–6100.e26 (2021). technology for cancer immunotherapy research. Front. Mol. Biosci. 7, 612801 (2020).
77. Michie, J. et al. Antagonism of IAPs enhances CAR T-cell efficacy. Cancer Immunol. Res. 113. Spitzer, M. H. & Nolan, G. P. Mass cytometry: single cells, many features. Cell 165,
7, 183–192 (2019). 780–791 (2016).
78. Giuffrida, L. et al. CRISPR/Cas9 mediated deletion of the adenosine A2A receptor enhances 114. Chong, E. A. et al. Pembrolizumab for B-cell lymphomas relapsing after or refractory to
CAR T cell efficacy. Nat. Commun. 12, 3236 (2021). CD19-directed CAR T-cell therapy. Blood 139, 1026–1038 (2022).
79. Papalexi, E. et al. Single-cell metabolic profiling of human cytotoxic T cells. Nature 12, 115. Goldberg, L. et al. Single-cell analysis by mass cytometry reveals CD19 CAR T cell
175–188 (2021). spatiotemporal plasticity in patients. Oncoimmunology 11, 2040772 (2022).
80. Mineo, M. et al. Tumor interferon signaling is regulated by a lncRNA INCR1 transcribed 116. Fisher, J. et al. Engineering γδT cells limits tonic signaling associated with chimeric
from the PD-L1 locus. Mol. Cell 78, 1207–1223.e8 (2020). antigen receptors. Sci. Signal. 12, eaax1872 (2019).
81. Sotillo, E. et al. Convergence of acquired mutations and alternative splicing of CD19 117. Rossi, J. et al. Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells are
enables resistance to CART-19 immunotherapy. Cancer Discov. 5, 1282–1295 (2015). associated with clinical outcomes in NHL. Blood 132, 804–814 (2018).
82. Pan, Y. et al. RNA dysregulation: an expanding source of cancer immunotherapy targets. 118. Bai, Z., Su, G. & Fan, R. Single-cell analysis technologies for immuno-oncology
Trends Pharmacol. Sci. 42, 268–282 (2021). research: from mechanistic delineation to biomarker discovery. Genomics Proteomics
83. Li, N. et al. CAR T cells targeting tumor-associated exons of glypican 2 regress Bioinformatics 19, 191–207 (2021).
neuroblastoma in mice. Cell Rep. Med. 2, 100297 (2021). 119. Patti, G. J., Yanes, O. & Siuzdak, G. Metabolomics: the apogee of the omics trilogy.
84. Tang, F. et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 6, Nat. Rev. Mol. Cell Biol. 13, 263–269 (2012).
377–382 (2009). 120. Kishton, R. J., Sukumar, M. & Restifo, N. P. Metabolic regulation of T cell longevity and
85. Armingol, E., Officer, A., Harismendy, O. & Lewis, N. E. Deciphering cell–cell interactions function in tumor immunotherapy. Cell Metab. 26, 94–109 (2017).
and communication from gene expression. Nat. Rev. Genet. 22, 71–88 (2021). 121. Fultang, L. et al. Metabolic engineering against the arginine microenvironment enhances
86. Suvà, M. L. & Tirosh, I. Single-cell RNA sequencing in cancer: lessons learned and CAR-T cell proliferation and therapeutic activity. Blood 136, 1155–1160 (2020).
emerging challenges. Mol. Cell 75, 7–12 (2019). 122. Hao, M. et al. Combination of metabolic intervention and T cell therapy enhances solid
87. Boroughs, A. C. et al. A distinct transcriptional program in human CAR T cells bearing the tumor immunotherapy. Sci. Transl. Med. 12, eaaz6667 (2020).
4-1BB signaling domain revealed by scRNA-seq. Mol. Ther. 28, 2577–2592 (2020). 123. Hirabayashi, K. et al. Dual-targeting CAR-T cells with optimal co-stimulation and
88. Li, X. X. et al. Single-cell transcriptomic analysis reveals BCMA CAR-T cell dynamics in a metabolic fitness enhance antitumor activity and prevent escape in solid tumors.
patient with refractory primary plasma cell leukemia. Mol. Ther. 29, 645–657 (2021). Nat. Cancer 2, 904–918 (2021).
89. Parker, K. R. et al. Single-cell analyses identify brain mural cells expressing CD19 as 124. Vardhana, S. A. et al. Impaired mitochondrial oxidative phosphorylation limits the
potential off-tumor targets for CAR-T immunotherapies. Cell 183, 126–142.e17 (2020). self-renewal of T cells exposed to persistent antigen. Nat. Immunol. 21, 1022–1033
90. Jing, Y. et al. Expression of chimeric antigen receptor therapy targets detected by (2020).
single-cell sequencing of normal cells may contribute to off-tumor toxicity. Cancer Cell 125. Gemta, L. F. et al. Impaired enolase 1 glycolytic activity restrains effector functions
39, 1558–1559 (2021). of tumor-infiltrating CD8+ T cells. Sci. Immunol. 4, eaap9520 (2019).
91. Boulch, M. et al. A cross-talk between CAR T cell subsets and the tumor microenvironment 126. Shen, L. et al. Metabolic reprogramming by ex vivo glutamine inhibition endows CAR-T
is essential for sustained cytotoxic activity. Sci. Immunol. 6, eabd4344 (2021). cells with less-differentiated phenotype and persistent antitumor activity. Cancer Lett.
92. Dhodapkar, K. M. et al. Changes in bone marrow tumor and immune cells correlate 538, 215710 (2022).
with durability of remissions following BCMA CAR T therapy in myeloma. Blood Cancer 127. Xu, J. et al. Metabolic flux analysis and fluxomics-driven determination of reaction free
Discov. https://doi.org/10.1158/2643-3230.BCD-22-0018 (2022). energy using multiple isotopes. Curr. Opin. Biotechnol. 64, 151–160 (2020).
93. Gohil, S. H., Iorgulescu, J. B., Braun, D. A., Keskin, D. B. & Livak, K. J. Applying high- 128. Liang, L., Sun, F., Wang, H. & Hu, Z. Metabolomics, metabolic flux analysis and cancer
dimensional single-cell technologies to the analysis of cancer immunotherapy. Nat. Rev. pharmacology. Pharmacol. Ther. 224, 107827 (2021).
Clin. Oncol. 18, 244–256 (2021). 129. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism
94. Bradley, P. & Thomas, P. G. Using T cell receptor repertoires to understand the principles pathways and impacts memory development in CAR T cells. Immunity 44, 380–390
of adaptive immune recognition. Annu. Rev. Immunol. 37, 547–570 (2019). (2016).
95. Pai, J. A. & Satpathy, A. T. High-throughput and single-cell T cell receptor sequencing 130. Van Bruggen, J. A. C. et al. Chronic lymphocytic leukemia cells impair mitochondrial
technologies. Nat. Methods 18, 881–892 (2021). fitness in CD8+ T cells and impede CAR T-cell efficacy. Blood 134, 44–58 (2019).
96. Sheih, A. et al. Clonal kinetics and single-cell transcriptional profiling of CAR-T cells in 131. Luo, M. et al. Race is a key determinant of the human intratumor microbiome. Cancer Cell
patients undergoing CD19 CAR-T immunotherapy. Nat. Commun. 11, 219 (2020). https://doi.org/10.1016/j.ccell.2022.08.007 (2022).
97. Melenhorst, J. J. et al. Decade-long leukaemia remissions with persistence of CD4+ CAR 132. Sepich-Poore, G. D. et al. The microbiome and human cancer. Science 371, eabc4552
T cells. Nature 602, 503–509 (2022). (2021).
98. Mani, D. R. et al. Cancer proteogenomics: current impact and future prospects. 133. Vétizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut
Nat. Rev. Cancer 22, 298–313 (2022). microbiota. Science 350, 1079–1084 (2015).
99. Enblad, G. et al. A phase I/IIa trial using CD19-targeted third-generation CAR T cells for 134. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against
lymphoma and leukemia. Clin. Cancer Res. 24, 6185–6194 (2018). epithelial tumors. Science 359, 91–97 (2018).
100. Hu, Z. et al. The Cancer Surfaceome Atlas integrates genomic, functional and drug 135. Sivan, A. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates
response data to identify actionable targets. Nat. Cancer 2, 1406–1422 (2021). anti-PD-L1 efficacy. Science 350, 1084–1089 (2015).
101. Chong, C., Coukos, G. & Bassani-Sternberg, M. Identification of tumor antigens with 136. Spencer, C. N. et al. Dietary fiber and probiotics influence the gut microbiome and
immunopeptidomics. Nat. Biotechnol. 40, 175–188 (2022). melanoma immunotherapy response. Science 374, 1632–1640 (2021).
102. Humphrey, S. J., Azimifar, S. B. & Mann, M. High-throughput phosphoproteomics reveals 137. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy
in vivo insulin signaling dynamics. Nat. Biotechnol. 33, 990–995 (2015). in melanoma patients. Science 359, 97–103 (2018).
103. Nix, M. A. et al. Surface proteomics reveals CD72 as a target for in vitro–evolved 138. Langille, M. G. I. et al. Predictive functional profiling of microbial communities using 16S
nanobody-based CAR-T cells in KMT2A/MLL1-rearranged B-ALL. Cancer Discov. 11, rRNA marker gene sequences. Nat. Biotechnol. 31, 814–821 (2013).
2032–2049 (2021). 139. Li, J. et al. An integrated catalog of reference genes in the human gut microbiome.
104. Perna, F. et al. Integrating proteomics and transcriptomics for systematic combinatorial Nat. Biotechnol. 32, 834–841 (2014).
chimeric antigen receptor therapy of AML. Cancer Cell 32, 506–519.e5 (2017). 140. Lloyd-Price, J. et al. Strains, functions and dynamics in the expanded Human Microbiome
105. Lee, J. K. et al. Systemic surfaceome profiling identifies target antigens for immune- Project. Nature 550, 61–66 (2017).
based therapy in subtypes of advanced prostate cancer. Proc. Natl Acad. Sci. USA 115, 141. Smith, M. et al. Gut microbiome correlates of response and toxicity following anti-CD19
E4473–E4482 (2018). CAR T cell therapy. Nat. Med. 28, 713–723 (2022).
106. Marcu, A. et al. HLA Ligand Atlas: a benign reference of HLA-presented peptides to 142. Matson, V., Chervin, C. S. & Gajewski, T. F. Cancer and the microbiome—influence
improve T-cell-based cancer immunotherapy. J. Immunother. Cancer 9, e002071 (2021). of the commensal microbiota on cancer, immune responses, and immunotherapy.
107. Salter, A. I. et al. Phosphoproteomic analysis of chimeric antigen receptor signaling Gastroenterology 160, 600–613 (2021).
reveals kinetic and quantitative differences that affect cell function. Sci. Signal. 11, 143. Moffitt, J. R., Lundberg, E. & Heyn, H. The emerging landscape of spatial profiling
eaat6753 (2018). technologies. Nat. Rev. Genet. https://doi.org/10.1038/s41576-022-00515-3 (2022).
108. Ramello, M. C. et al. An immunoproteomic approach to characterize the CAR 144. Ståhl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial
interactome and signalosome. Sci. Signal. 12, eaap9777 (2019). transcriptomics. Science 353, 78–82 (2016).

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 226


Review article

145. He, S. et al. High-plex imaging of RNA and proteins at subcellular resolution in fixed 181. Spiegel, J. Y. et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with
tissue by spatial molecular imaging. Nat. Biotechnol. 40, 1794–1806 (2022). recurrent or refractory B cell malignancies: a phase 1 trial. Nat. Med. 27, 1419–1431 (2021).
146. Longo, S. K., Guo, M. G., Ji, A. L. & Khavari, P. A. Integrating single-cell and spatial 182. Fernández de Larrea, C. et al. Defining an optimal dual-targeted CAR T-cell therapy
transcriptomics to elucidate intercellular tissue dynamics. Nat. Rev. Genet. 22, 627–644 approach simultaneously targeting BCMA and GPRC5D to prevent BCMA escape–driven
(2021). relapse in multiple myeloma. Blood Cancer Discov. 1, 146–154 (2020).
147. Deng, Y. et al. Spatial profiling of chromatin accessibility in mouse and human tissues. 183. Han, X. et al. Construction of a human cell landscape at single-cell level constructing an
Nature 609, 375–383 (2022). HCL using microwell-seq. Nature 581, 303–309 (2020).
148. Deng, Y. et al. Spatial-CUT&Tag: spatially resolved chromatin modification profiling at the 184. He, S. et al. Single-cell transcriptome profiling of an adult human cell atlas of 15 major
cellular level. Science 375, 681–686 (2022). organs. Genome Biol. 21, 294 (2020).
149. Giesen, C. et al. Highly multiplexed imaging of tumor tissues with subcellular resolution 185. Tabula Sapiens Consortium et al. The Tabula Sapiens: a multiple-organ, single-cell
by mass cytometry. Nat. Methods 11, 417–422 (2014). transcriptomic atlas of humans. Science 376, eabl4896 (2022).
150. Kuett, L. et al. Three-dimensional imaging mass cytometry for highly multiplexed 186. Eraslan, G. et al. Single-nucleus cross-tissue molecular reference maps to decipher
molecular and cellular mapping of tissues and the tumor microenvironment. Nat. Cancer disease gene function. Science 376, eabl4290 (2022).
3, 122–133 (2022). 187. Suo, C. et al. Mapping the developing human immune system across organs. Science
151. Jackson, H. W. et al. The single-cell pathology landscape of breast cancer. Nature 578, 376, eabo0510 (2022).
615–620 (2020). 188. Domínguez Conde, C. et al. Cross-tissue immune cell analysis reveals tissue-specific
152. Schulz, D. et al. Simultaneous multiplexed imaging of mRNA and proteins with subcellular features in humans. Science 376, eabl5197 (2022).
resolution in breast cancer tissue samples by mass cytometry. Cell Syst. 6, 25–36.e5 (2018). 189. Rozenblatt-Rosen, O. et al. The Human Tumor Atlas Network: charting tumor transitions
153. Hoch, T. et al. Multiplexed imaging mass cytometry of the chemokine milieus in across space and time at single-cell resolution. Cell 181, 236–249 (2020).
melanoma characterizes features of the response to immunotherapy. Sci. Immunol. 7, 190. Bausch-Fluck, D. et al. The in silico human surfaceome. Proc. Natl Acad. Sci. USA 115,
eabk1692 (2022). E10988–E10997 (2018).
154. Hartmann, F. J. et al. Single-cell metabolic profiling of human cytotoxic T cells. 191. Chen, N., Li, X., Chintala, N. K., Tano, Z. E. & Adusumilli, P. S. Driving CARs on the uneven
Nat. Biotechnol. 39, 186–197 (2021). road of antigen heterogeneity in solid tumors. Curr. Opin. Immunol. 51, 103–110 (2018).
155. Lee, W. D., Mukha, D., Aizenshtein, E. & Shlomi, T. Spatial-fluxomics provides a 192. Arcangeli, S. et al. CAR T cell manufacturing from naive/stem memory T lymphocytes
subcellular-compartmentalized view of reductive glutamine metabolism in cancer cells. enhances antitumor responses while curtailing cytokine release syndrome. J. Clin. Invest.
Nat. Commun. 10, 1351 (2019). 132, e150807 (2022).
156. Avila Cobos, F., Alquicira-Hernandez, J., Powell, J. E., Mestdagh, P. & De Preter, K. 193. Turtle, C. J. et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL
Benchmarking of cell type deconvolution pipelines for transcriptomics data. patients. J. Clin. Invest. 126, 2123–2138 (2016).
Nat. Commun. 11, 5650 (2020). 194. Bai, Z. et al. Single-cell antigen-specific landscape of CAR T infusion product identifies
157. Tran, H. T. N. et al. A benchmark of batch-effect correction methods for single-cell RNA determinants of CD19-positive relapse in patients with ALL. Sci. Adv. 8, eabj2820 (2022).
sequencing data. Genome Biol. 21, 12 (2020). 195. Larson, R. C. et al. CAR T cell killing requires the IFNγR pathway in solid but not liquid
158. Kiselev, V. Y., Andrews, T. S. & Hemberg, M. Challenges in unsupervised clustering tumours. Nature 604, 563–570 (2022).
of single-cell RNA-seq data. Nat. Rev. Genet. 20, 273–282 (2019). 196. Seo, H. et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR
159. Black, J. R. M. & McGranahan, N. Genetic and non-genetic clonal diversity in cancer T cells. Nat. Immunol. 22, 983–995 (2021).
evolution. Nat. Rev. Cancer 21, 379–392 (2021). 197. Tang, J. P. et al. Hypophosphatemia due to increased effector cell metabolic activity
160. Orlando, E. J. et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute is associated with neurotoxicity symptoms in CD19-targeted CAR T-cell therapy.
lymphoblastic leukemia. Nat. Med. 24, 1504–1506 (2018). Cancer Immunol. Res. 10, 1433–1440 (2022).
161. Sammut, S.-J. et al. Multi-omic machine learning predictor of breast cancer therapy 198. Nobles, C. L. et al. CD19-targeting CAR T cell immunotherapy outcomes correlate with
response. Nature 601, 623–629 (2022). genomic modification by vector integration. J. Clin. Invest. 130, 673–685 (2020).
162. Kang, M., Ko, E. & Mersha, T. B. A roadmap for multi-omics data integration using deep 199. Cappell, K. M. & Kochenderfer, J. N. A comparison of chimeric antigen receptors
learning. Brief. Bioinform. 23, bbab454 (2022). containing CD28 versus 4-1BB costimulatory domains. Nat. Rev. Clin. Oncol. 18, 715–727
163. Goecks, J., Jalili, V., Heiser, L. M. & Gray, J. W. How machine learning will transform (2021).
biomedicine. Cell 181, 92–101 (2020). 200. Lemoine, J., Ruella, M. & Houot, R. Born to survive: how cancer cells resist CAR T cell
164. Miao, Z., Humphreys, B. D., McMahon, A. P. & Kim, J. Multi-omics integration in the age therapy. J. Hematol. Oncol. 14, 199 (2021).
of million single-cell data. Nat. Rev. Nephrol. 17, 710–724 (2021). 201. Lemoine, J., Ruella, M. & Houot, R. Overcoming intrinsic resistance of cancer cells to CAR
165. Wei, J., Han, X., Bo, J. & Han, W. Target selection for CAR-T therapy. J. Hematol. Oncol. 12, T-cell killing. Clin. Cancer Res. 27, 6298–6306 (2021).
62 (2019). 202. Plaks, V. et al. CD19 target evasion as a mechanism of relapse in large B-cell lymphoma
166. Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and treated with axicabtagene ciloleucel. Blood 138, 1081–1085 (2021).
functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021). 203. Rabilloud, T. et al. Single-cell profiling identifies pre-existing CD19-negative subclones
167. Bosse, K. R. et al. Identification of GPC2 as an oncoprotein and candidate in a B-ALL patient with CD19-negative relapse after CAR-T therapy. Nat. Commun. 12, 865
immunotherapeutic target in high-risk neuroblastoma. Cancer Cell 32, 295–309.e12 (2017). (2021).
168. Yarmarkovich, M. et al. Cross-HLA targeting of intracellular oncoproteins with 204. Asnani, M. et al. Retention of CD19 intron 2 contributes to CART-19 resistance in
peptide-centric CARs. Nature 599, 477–484 (2021). leukemias with subclonal frameshift mutations in CD19. Leukemia 34, 1202–1207 (2020).
169. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass 205. Jacoby, E. et al. CD19 CAR immune pressure induces B-precursor acute lymphoblastic
spectrometry and exome sequencing. Nature 515, 572–576 (2014). leukaemia lineage switch exposing inherent leukaemic plasticity. Nat. Commun. 7, 12320
170. Hsiue, E. H. C. et al. Targeting a neoantigen derived from a common TP53 mutation. (2016).
Science 371, eabc8697 (2021). 206. Gardner, R. et al. Acquisition of a CD19-negative myeloid phenotype allows immune
171. Thorsson, V. et al. The immune landscape of cancer. Immunity 48, 812–830.e14 (2018). escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 127, 2406–2410
172. Wu, J. et al. TSNAdb: a database for tumor-specific neoantigens from immunogenomics (2016).
data analysis. Genomics Proteomics Bioinformatics 16, 276–282 (2018). 207. Ruella, M. et al. Induction of resistance to chimeric antigen receptor T cell therapy by
173. Dannenfelser, R. et al. Discriminatory power of combinatorial antigen recognition in transduction of a single leukemic B cell. Nat. Med. 24, 1499–1503 (2018).
cancer T cell therapies. Cell Syst. 11, 215–228.e5 (2020). 208. Upadhyay, R. et al. A critical role for FAS-mediated off-target tumor killing in T-cell
174. Kloss, C. C., Condomines, M., Cartellieri, M., Bachmann, M. & Sadelain, M. Combinatorial immunotherapy. Cancer Discov. 11, 599–613 (2021).
antigen recognition with balanced signaling promotes selective tumor eradication by 209. Monti, S. et al. Integrative analysis reveals an outcome-associated and targetable pattern
engineered T cells. Nat. Biotechnol. 31, 71–75 (2013). of p53 and cell cycle deregulation in diffuse large B cell lymphoma. Cancer Cell 22,
175. Roybal, K. T. et al. Engineering T cells with customized therapeutic response programs 359–372 (2012).
using synthetic Notch receptors. Cell 167, 419–432.e16 (2016). 210. Jain, M. D. et al. Tumor interferon signaling and suppressive myeloid cells are associated
176. Fedorov, V. D., Themeli, M. & Sadelain, M. PD-1- and CTLA-4-based inhibitory chimeric with CAR T-cell failure in large B-cell lymphoma. Blood 137, 2621–2633 (2021).
antigen receptors (iCARs) divert off-target immunotherapy responses. Sci. Transl. Med. 211. Kankeu Fonkoua, L. A., Sirpilla, O., Sakemura, R., Siegler, E. L. & Kenderian, S. S. CAR T
5, 215ra172 (2013). cell therapy and the tumor microenvironment: current challenges and opportunities.
177. Cho, J. H., Collins, J. J. & Wong, W. W. Universal chimeric antigen receptors for Mol. Ther. Oncolytics 25, 69–77 (2022).
multiplexed and logical control of T cell responses. Cell 173, 1426–1438.e11 (2018). 212. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for
178. Cho, J. H. et al. Engineering advanced logic and distributed computing in human CAR effective therapy. Nat. Med. 24, 541–550 (2018).
immune cells. Nat. Commun. 12, 792 (2021). 213. Britten, C. M., Shalabi, A. & Hoos, A. Industrializing engineered autologous T cells as
179. Atilla, P. A. et al. Combinatorial antigen targeting strategies for acute leukemia: medicines for solid tumours. Nat. Rev. Drug Discov. 20, 476–488 (2021).
application in myeloid malignancy. Cytotherapy 24, 282–290 (2022). 214. Xu, N. et al. STING agonist promotes CAR T cell trafficking and persistence in breast
180. Liu, F. et al. First-in-human CLL1-CD33 compound CAR T cell therapy induces complete cancer. J. Exp. Med. 218, e20200844 (2021).
remission in patients with refractory acute myeloid leukemia: update on phase 1 clinical 215. Zhang, X. et al. Dissecting esophageal squamous-cell carcinoma ecosystem by single-cell
trial. Blood 132, 901–901 (2018). transcriptomic analysis. Nat. Commun. 12, 5291 (2021).

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 227


Review article

216. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 239. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy
374, abe6474 (2021). response. Nature 577, 549–555 (2020).
217. Adusumilli, P. S. et al. A phase I trial of regional mesothelin-targeted CAR T-cell therapy 240. Jing, Y., Yang, J., Johnson, D. B., Moslehi, J. J. & Han, L. Harnessing big data to
in patients with malignant pleural disease, in combination with the anti-PD-1 agent characterize immune-related adverse events. Nat. Rev. Clin. Oncol. 19, 269–280 (2022).
pembrolizumab. Cancer Discov. 11, 2748–2763 (2021). 241. Reel, P. S., Reel, S., Pearson, E., Trucco, E. & Jefferson, E. Using machine learning
218. Rafiq, S. et al. Targeted delivery of a PD-1-blocking scFV by CAR-T cells enhances anti- approaches for multi-omics data analysis: a review. Biotechnol. Adv. 49, 107739
tumor efficacy in vivo. Nat. Biotechnol. 36, 847–858 (2018). (2021).
219. Cherkassky, L. et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade 242. Levine, L. S. et al. Single-cell analysis by mass cytometry reveals metabolic states
resist tumor-mediated inhibition. J. Clin. Invest. 126, 3130–3144 (2016). of early-activated CD8+ T cells during the primary immune response. Immunity 54,
220. Choi, B. D. et al. CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII 829–844.e5 (2021).
CAR T cells in a preclinical model of human glioblastoma. J. Immunother. Cancer 7, 304
(2019).
Acknowledgements
221. Yamamoto, T. N. et al. T cells genetically engineered to overcome death signaling
The work of the authors is supported by the US NIH (grants R01HG011633 and R01CA262623
enhance adoptive cancer immunotherapy. J. Clin. Invest. 129, 1551–1565 (2019).
to L.H.). The content is solely the responsibility of the authors and does not necessarily
222. Avanzi, M. P. et al. Engineered tumor-targeted T cells mediate enhanced anti-tumor
represent the official views of the NIH. M.R.G. is a Scholar of the Leukaemia and Lymphoma
efficacy both directly and through activation of the endogenous immune system.
Society. The authors regret that page limitations have prevented them from including
Cell Rep. 23, 2130–2141 (2018).
all the relevant studies in this Review. Draft figures for this manuscript were created with
223. Luu, M. et al. Microbial short-chain fatty acids modulate CD8+ T cell responses and
BioRender.com.
improve adoptive immunotherapy for cancer. Nat. Commun. 12, 4077 (2021).
224. Ellis, G. I., Sheppard, N. C. & Riley, J. L. Genetic engineering of T cells for immunotherapy.
Nat. Rev. Genet. 22, 427–447 (2021). Author contributions
225. Brudno, J. N. & Kochenderfer, J. N. Recent advances in CAR T-cell toxicity: mechanisms, All authors made a substantial contribution to all aspects of the preparation of this manuscript.
manifestations and management. Blood Rev. 34, 45–55 (2019).
226. Kennedy, L. B. & Salama, A. K. S. A review of cancer immunotherapy toxicity.
Competing interests
CA Cancer J. Clin. 70, 86–104 (2020).
M.R.G. has stock ownership interest in KDAc Therapeutics, receives funding from Abbvie,
227. Dougan, M., Luoma, A. M., Dougan, S. K. & Wucherpfennig, K. W. Understanding and treating
Allogene, Kite/Gilead and Sanofi, and has received honoraria from Tessa Therapeutics. J.Y.,
the inflammatory adverse events of cancer immunotherapy. Cell 184, 1575–1588 (2021).
Y.C., Y.J. and L.H. declare no competing interests.
228. Morris, E. C., Neelapu, S. S., Giavridis, T. & Sadelain, M. Cytokine release syndrome and
associated neurotoxicity in cancer immunotherapy. Nat. Rev. Immunol. 22, 85–96 (2022).
229. Teachey, D. T. et al. Identification of predictive biomarkers for cytokine release syndrome Additional information
after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Correspondence should be addressed to Michael R. Green or Leng Han.
Cancer Discov. 6, 664–679 (2016).
230. Gill, S. & Brudno, J. N. CAR T-cell therapy in hematologic malignancies: clinical role, Peer review information Nature Reviews Clinical Oncology thanks M. Maus, who co-reviewed
toxicity, and unanswered questions. Am. Soc. Clin. Oncol. Educ. B 41, e246–e265 with N. Haradhvala; R. Fan, who co-reviewed with Z. Bai; and the other, anonymous, reviewer(s)
(2021). for their contribution to the peer review of this work.
231. Depil, S., Duchateau, P., Grupp, S. A., Mufti, G. & Poirot, L. ‘Off-the-shelf’ allogeneic CAR
T cells: development and challenges. Nat. Rev. Drug Discov. 19, 185–199 (2020). Reprints and permissions information is available at www.nature.com/reprints.
232. Bai, Z. et al. Single-cell multiomics dissection of basal and antigen-specific activation
states of CD19-targeted CAR T cells. J. Immunother. Cancer 9, e002328 (2021). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
233. Davis-Marcisak, E. F. et al. From bench to bedside: single-cell analysis for cancer published maps and institutional affiliations.
immunotherapy. Cancer Cell 39, 1062–1080 (2021).
234. Gawad, C., Koh, W. & Quake, S. R. Single-cell genome sequencing: current state of the Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
science. Nat. Rev. Genet. 17, 175–188 (2016). article under a publishing agreement with the author(s) or other rightsholder(s); author self-
235. Andrews, T. S., Kiselev, V. Y., McCarthy, D. & Hemberg, M. Tutorial: guidelines for the archiving of the accepted manuscript version of this article is solely governed by the terms of
computational analysis of single-cell RNA sequencing data. Nat. Protoc. 16, 2749–2764 such publishing agreement and applicable law.
(2021).
236. Guo, S., Zhang, C. & Le, A. The limitless applications of single-cell metabolomics.
Curr. Opin. Biotechnol. 71, 115–122 (2021). Related links
237. Lloréns-Rico, V., Simcock, J. A., Huys, G. R. B. & Raes, J. Single-cell approaches in human Antigen Explorer: https://antigen.princeton.edu/
microbiome research. Cell 185, 2725–2738 (2022). CARTSC: https://hanlab.tamhsc.edu/CARTSC/
238. Johnson, D. B. et al. Quantitative spatial profiling of PD-1/PD-L1 interaction and HLA-DR/ ClinicalTrials.gov: https://clinicaltrials.gov/ct2/home
IDO-1 predicts improved outcomes of anti-PD-1 therapies in metastatic melanoma.
Clin. Cancer Res. 24, 5250–5260 (2018). © Springer Nature Limited 2023

Nature Reviews Clinical Oncology | Volume 20 | April 2023 | 211–228 228

You might also like