1..23 ++
1..23 ++
1..23 ++
Abstract
Keywords
Allogeneic cancer therapy · Cancer genetic engineering · Cancer
immunotherapy · Chimeric antigen receptor (CAR) · CRISPR-Cas9 ·
Graft-versus-host disease (GvHD) · Natural killer T (NKT) cell · Stem cell
1 Introduction
The pursuit of innovative cancer therapies and stem cells has emerged as a promising
avenue of exploration. This section delves into the various types of stem cells—
embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal
stem cells (MSCs), and hematopoietic stem cells (HSCs)—as potential cell sources
for advancing cancer treatment (Sagar et al. 2007). Each stem cell type possesses
unique attributes and faces distinct challenges within the context of cancer therapy.
ESCs represent a unique class of pluripotent cells derived from embryos fertilized
in vitro. The process of ESC cultivation involves the development of these embryos
into blastocysts, from which the undifferentiated inner cell mass (ICM) can be
isolated. The hallmark characteristic of ESCs is their pluripotency, enabling them
to self-renew indefinitely and differentiate into all cell types except for placenta
(Hochedlinger and Jaenisch 2003; Wobus and Boheler 2005). This inherent versa-
tility positions ESCs as an attractive candidate for cancer treatment and development
Stem Cell-Derived Cell Therapy for Cancer
of tumor models. ESCs can be programmed into therapeutic cells, particularly T and
natural killer (NK) cells, which can be genetically modified to express tumor-
suppressing proteins, immunostimulatory molecules, and chimeric antigen receptors
(CARs), enabling targeted cancer therapy and immune cell recruitment to tumor sites
(Hochedlinger and Jaenisch 2003; Wobus and Boheler 2005; Zeng et al. 2012).
These customized cells can effectively target cancer cells or recruit other immune
elements to the tumor microenvironment, fostering a multifaceted approach to
cancer treatment. However, the extensive proliferative and differentiating
capabilities of ESCs present potential drawbacks. Their propensity for unlimited
division mirrors cancer behavior, raising concerns about tumorigenic potential,
especially following genetic modifications. Ethical dilemmas also loom over
ESCs, as their generation involves the destruction of in vitro-fertilized embryos,
sparking regulatory variations across countries and affecting ESC research (Tam
et al. 2023; Iltis et al. 2023). Overall, the complex signaling pathways governing
ESC behavior remain incompletely understood, and overcoming immune rejection
and generating safe, heterogeneous cell lines pose ongoing challenges in ESC-based
therapies (Fig. 1).
iPSCs share functional similarities with ESCs while diverging in their origin,
derived through reprogramming adult somatic cells, such as dermal fibroblasts,
peripheral blood cells, or adipose tissue cells. This reprogramming process involves
Fig. 1 Stem cell sources for cell therapy. A diverse array of stem cell sources suitable for cell
therapy is shown, encompassing embryonic stem cells, induced pluripotent stem cells, mesenchy-
mal stem cells, and hematopoietic stem cells, among others.
M. Wilson et al.
the activation of key pluripotency factors like Oct4, Sox2, and Klf4 (Sharkis et al.
2012; Hansen et al. 2018). iPSCs closely mimic ESCs in research settings, providing
an ethical advantage by utilizing cells sourced from consenting adult donors. This
ethical consideration facilitates their transition into clinical trials, where iPSCs
exhibit potential in cancer therapy, particularly in cell-based immunotherapy
(Li et al. 2018; Gutbier et al. 2020; Frank et al. 2020; Iriguchi et al. 2021; Aoki
et al. 2023). They can be reprogrammed into T lymphocytes, retaining T-cell
receptor genes for subsequent differentiation into T cells. These reprogrammed
T cells can be engineered to express CARs and other molecules for targeted cancer
therapy (Li et al. 2018; Allen et al. 2021). iPSCs’ ability to differentiate into various
tissue and cell types offers opportunities for tissue regeneration and disease
modeling, aiding diagnostics, and advancing therapeutics. Nevertheless, the devel-
opment of effective differentiation protocols for desired cell types from iPSCs
remains labor-intensive due to their diverse sources and potential differentiations.
The absence of Good Manufacturing Practice (GMP)-compliant protocols raises
safety concerns during iPSC generation, expansion, and differentiation, emphasizing
the need for standardized procedures to ensure consistency within clinical practice.
Another promising source for cancer therapy resides in MSCs, originating from
bone marrow, adipose tissue, and umbilical cord tissue. MSCs possess the ability
to differentiate into bone, cartilage, and fat tissues, and they are noted for their
regenerative and immunomodulatory properties (Hmadcha et al. 2020). These
attributes render MSCs suitable for clinical trials spanning orthopedic injuries,
autoimmune diseases, and neurological disorders. Beyond their conventional stem-
cell traits, such as self-renewal and robust differentiation capabilities, MSCs exhibit
a unique tumor-homing phenomenon. Driven by tumor tropism, chemotaxis, and the
tumor microenvironment, MSCs are naturally attracted to tumor sites, where they
engage with cancer cells, contributing to the attack against cancerous cells (Shah
2012; Datta et al. 2022). Genetic modifications further enhance their therapeutic
potential by enabling MSCs to express exogenous tumor-targeting or therapeutic
molecules (Shah 2012; Datta et al. 2022; Yan et al. 2023). Additionally, their low
immunogenicity, attributed to minimal MHC-I molecule expression and immuno-
modulatory capabilities, renders MSC allogeneic transplantation well-tolerated. The
duality of MSCs’ interactions with the tumor microenvironment may pose a chal-
lenge. Despite interrupting cancer cell replication to inhibit tumor growth, MSCs can
also differentiate into cancer-associated fibroblasts (CAFs), potentially contributing
to cancer progression and immunosuppression (Mishra et al. 2009; Datta et al.
2022). The outcome of these interactions depends on various factors, including the
tumor model, MSC source, timing, dosage of treatment, delivery methods, and
patient-specific conditions, complicating their application in clinical oncology.
HSCs offer distinct advantages as a potential cell source for cancer therapy,
owing to their natural involvement in blood and immune cell production. Their
integral role in immune cell generation underpins their application in cancer therapy,
notably in HSC transplantation (HSCT). HSCT facilitates reconstruction of a
patient’s immune system, providing a valuable tool for treating hematological
malignancies such as leukemia and lymphoma (Rowe et al. 2016; Ng and Alexander
Stem Cell-Derived Cell Therapy for Cancer
Stem cells possess the potential for genetic modifications and the ability to replicate
in a clonal manner, resulting in the generation of exceptionally pure effector cells.
Advancements in stem cell culture and differentiation techniques have broadened the
spectrum of stem cell-derived cellular candidates. This now encompasses conven-
tional T cells, non-conventional T cells, NK cells, dendritic cells (DCs),
macrophages, and myeloid cells, all of which can be further engineered to acquire
anticancer properties (Fig. 2).
The groundbreaking achievements of immune checkpoint inhibitors and CAR-T
cell therapies, exemplified by the FDA-approved anti-CD19 CAR-T cell therapy for
B-cell malignancies, have underscored the critical role of T cells in immunotherapy
(Sterner and Sterner 2021). Accordingly, conventional αβ T cells have emerged as a
pivotal component of the investigation of stem cell engineering. In alignment with
established T-cell genetic engineering techniques, stem cell-derived αβ T cells can
be modified to express physiological TCRs or synthetic CARs to acquire specificity
M. Wilson et al.
CAR-
CAR-engineering
αβ T iNKT NK
TCR - NK
engineering Differentiation
MAIT γδ T
CD16-engineering CD16-
NK
Stem
cells
CAR-engineering
CAR- TCR/CAR-
CAR-T
iNKT engineering Differentiation Macrophage
DC Myeloid
CAR- CAR-
MAIT γδ T
Fig. 2 Differentiated cell types for therapy. The figure shows various types of differentiated cells
that can be derived from stem cells for cancer therapy and how these differentiated cells can be
engineered to possess anticancer properties. iNKT invariant natural killer T cell, MAIT mucosal-
associated invariant T cell, NK natural killer cell, DC dendritic cell
for tumor antigens (Sadelain et al. 2017). For example, TCR-engineered T cells such
as NY-ESO-1-specific TCR-engineered T cells have propelled advancements in the
treatment for myeloma, lung cancer, melanoma, and sarcoma (Rapoport et al. 2015;
Zhang et al. 2022b). In addition to anti-CD19 CAR-T cell therapy, B-cell maturation
antigen (BCMA) CAR-T cell therapy has also gained approval for the treatment of
multiple myeloma and various B-cell malignancies (Raje et al. 2019; D’Agostino
and Raje 2020; Li et al. 2023). Stem cell-derived αβ T cells can further mitigate the
limitations of current CAR-T cell therapy, particularly cytokine release syndrome
(CRS), owing to their different cytokine patterns (Themeli et al. 2013; Li et al.
2023). Moreover, considering the clinical significance of the CD4+:CD8+ ratio in
therapeutic outcomes, allogeneic αβ T cells derived from induced pluripotent stem
cells (iPSCs) or hematopoietic stem cells (HSCs) can circumvent the challenges
posed by unstable or unfavorable T-cell compositions through precise gene editing
techniques (Themeli et al. 2015; Turtle et al. 2016).
NK cells distinguish target cells through a combination of activating and inhibi-
tory receptors, independently of MHC recognition, thereby mitigating the risk of
GvHD, a concern often associated with conventional CAR-T cell therapy (Ruggeri
et al. 2007; Fang et al. 2017; Rezvani et al. 2017; Abel et al. 2018). Similar to NK
cells derived from other sources ranging from autologous patients to allogeneic
Stem Cell-Derived Cell Therapy for Cancer
4 Mechanisms of Action
Watarai et al. demonstrated that upon α-GalCer stimulation, iPSC-iNKT cells can
expand in number, facilitate DC maturation, and activate CTLs and NK cells by
releasing IFN-γ (Watarai et al. 2010). In addition to IFN-γ secreted by iPSC-iNKT,
other pro-inflammatory/antitumor cytokines may be supplemented with a diverse
array of stem cell-derived immune cells in cancer immunotherapy. Upon
TCR-dependent activation, γδ T cells possess the capacity to release abundant
pro-inflammatory cytokines including IL-2, IL-12, IL-15, and IL-18, therefore
improving rapid, direct cell killing (Kabelitz et al. 2020). Similarly, MAIT cells,
characterized by their high expression of pro-inflammatory cytokine receptors such
as IL-12R and IL-18R, reciprocally secrete antitumor cytokines including IFN-γ and
TNF-α, which can be further complemented by stem cell-derived MAIT cells (Petley
et al. 2021). Moreover, stem cell-derived immune cells can be genetically modified
to express antitumor cytokines. For instance, Mizuhashi et al. reported that mouse
iPSC-derived proliferating MCs (iPSC-pMCs), genetically engineered to produce
IL-15 and IL-15/Rα, can augment the NK-mediated cytotoxicity in mice, improving
the treatment for mouse melanoma with major histocompatibility complex (MHC)
class I loss (Mizuhashi et al. 2021).
5 Engineering Approaches
Within the field of cancer treatment, cell-based therapeutics hold great potential, but
are limited by several factors, including high variability, protracted manufacturing,
high sensitivity and low stability, and product heterogeneity (Taefehshokr et al.
2022; Blache et al. 2022). Stringent GMP regulations may abate variability between
batches of cell products by standardizing cell characterization, including testing of
yield, purity, and tumorigenicity (Bedford et al. 2018). Additionally, external aspects
to the manufacturing process itself may impact efficacy in the clinic, including stem
cell origin, administration route, dosing, and combination with other products, to
name a few.
The preclinical applications of these cell products have yielded promising results in
the treatment of a wide range of cancers across various models. Among the most well-
explored preclinically are DCs produced from stem cells as a vaccine to stimulate an
immune response through cancer antigen presentation. Engineering can further facili-
tate DC enhancement either through self-expression of target cancer antigens or
overexpression of presenting molecules (i.e., MHC-I and II, CD1d, MR1, etc.) to
promote stimulation of endogenous lymphocytes (Zeng et al. 2012; Tominaga et al.
2023). Regarding cytotoxic cell types, NK and T cells have been the most extensively
stem cell-derived therapeutic cells. Belonging to opposite arms of innate and adaptive
immunity, both employ unique recognition and killing pathways that may be homed to
tumor cells using CAR engineering (Huang et al. 2022; Klaihmon et al. 2023).
Through innate-like T cells, which simultaneously express NK activating markers
and a lineage-specific TCR, the antitumor potential of both cell types may be
synergized within a single model. In our hands, we have developed a malleable
platform for engineering CAR-iNKT cells against a variety of tumor-specific antigens,
showing strong suppression of cancer growth with limited GvHD onset in mice
(Li et al. 2021c, 2022b). The potency of innate-like T cells has yet to be fully explored
in the preclinical setting, providing potential opportunities to expand upon the
established success of CAR-T cell therapy. Although therapies encompassing other
unconventional T cells, such as MAIT and γδ T cells, have not yet broken through to
clinical trials, they still show great promise within the preclinical setting (Wakao et al.
2013; Watanabe et al. 2018; Zeng et al. 2019). Through a stem cell-based platform,
rare immune cell subtypes such as these may be more easily generated for stockpiling
of “off-the-shelf” therapeutic doses for cancer patients.
Expanding on this preclinical success, several clinical trials have been established
to examine the safety and potency of stem cell-based therapies in cancer patients
(Table 1). While the majority of these studies are still recruiting or remain ongoing,
those that have concluded have shown promising results for patient remission. In
particular, Fate Therapeutics has established several therapeutic CAR-NK cell
products derived from a human clonal master iPSC line, providing both a highly
malleable and controlled platform for generating therapeutic cells. FT500, their
earliest CAR-NK cell product, has demonstrated strong preclinical potency,
engaging in innate cytotoxicity, producing adjuvant stimulation of endogenous
T cells, and augmenting the efficacy of anti-PD-1 combination therapy (Cichocki
Table 1 Current stem cell-derived cell therapies in preclinical or clinical trials
Clinical trial
Cell therapy Phase identifier Description Results
NK Clinical NCT05182073 FT576 in Subjects with Multiple Myeloma Phase I: Recruiting
NCT05336409 CNTY-101 in Participants with CD19+ B-Cell Phase I: Recruiting
Malignancies
NCT04630769 FT516 and IL2 with Enoblituzumab for Ovarian Phase I: Completed—Limited adverse systemic
NCT04551885 Cancer; effects reported in all individuals;
NCT04023071 Solid Tumors; Phase I: Active;
Hematologic Malignancies Phase I: Active
NCT05950334 FT522 With Rituximab in Relapsed B-Cell Phase I: Not Yet Recruiting
Lymphoma
NCT05069935; FT538 in Combination with mAb in Advanced Phase I: Active;
NCT04714372; Solid Tumors; Phase I: Recruiting;
NCT04614636; Acute Myeloid Leukemia; Phase I: Active;
NCT05708924 Advanced Hematologic Malignancies; Phase I: Recruiting
Recurrent Ovarian, Fallopian Tube, Primary
Peritoneal Cancer
NCT05395052 FT536 Monotherapy and Combination with mAb Phase I: Active
in Advanced Solid Tumors
NCT04245722; FT596 as Monotherapy and Combination with anti- Phase I: Active;
NCT04555811 CD20 mAb; Phase I: Active
Combination with Rituximab for Relapse
Prevention in Non-Hodgkin Lymphoma
NCT03841110; FT500 as Monotherapy and Combination with Phase I: Results not yet reported;
NCT04106167 Immune Checkpoint Inhibitors in Solid Tumor; Ongoing
Long-term, Observational Study Post-Treatment
Preclinical iPSC-CD19CAR NK cells show selective targeting of CD19+ hematologic cancers in vitro and elevated NK activation marker
levels post-exposure (Klaihmon et al. 2023; Chang et al. 2023)
iPSC-MCAR NK cells display enhanced cytotoxicity against solid TNBC in vitro and in vivo (Yang et al. 2023a)
M. Wilson et al.
DC Clinical Not Available Therapeutic DC Vaccination with IL-2 as Phase I/II Trial: Autologous HSC-derived DCs
Consolidation Therapy for Ovarian Cancer Patients secrete IL-12, reduce inhibitory CD4+CD25+
T cell subsets, and enhance proportion of CD8+
IFNγ-secreting T cells (Baek et al. 2011)
Preclinical Embryoid bodies form mixed CD11c+ DC populations, which can be enriched for cross-presentation specialized CD141+ DCs
through CD1c+ depletion (Sachamitr et al. 2017)
iPSCs (Horton et al. 2020; Oba et al. 2021; Tominaga et al. 2023), ESCs (Zeng et al. 2012), and CBSCs (Chang et al. 2012) are
viable input for generation of stem-cell-derived DCs, which show strong adjuvant stimulation and cytokine profile
Adjuvant effects can be further enhanced through genetic engineering of tumor antigens expression (e.g., MSLN) (Tominaga
et al. 2023) or overexpression of antigen-presenting markers (e.g., CD1d) (Zeng et al. 2012)
Macrophage Preclinical iPSC-macrophages show proper function and polarization compared to primary macrophages (Gutbier et al. 2020)
iPSC-CD19CAR macrophages display durable suppression of K562 leukemia cells in vitro and in vivo (Zhang et al. 2020)
T Clinical NCT04629729 FT819 in Subjects with B-Cell Malignancies Phase I: Recruiting
Stem Cell-Derived Cell Therapy for Cancer
Preclinical iPSC-T cells display normal differentiation into single-positive lineages (Nishimura and Nakauchi 2019; Iriguchi et al. 2021)
CD8 αβ T cells demonstrate transient ability to suppress H226 hepatocarcinoma growth in vitro and in vivo (Iriguchi et al. 2021)
Anti-EGFRvIII iPSC-CAR T cells demonstrate extravasation across the blood-brain barrier and significantly reduce within
GBM burden in vitro (Huang et al. 2022)
iNKT Clinical jRCT2033200116 Phase I Study of iPS-NKT in Patients with Phase I: Ongoing
Recurrent or Advanced Head and Neck Cancer
Preclinical iPSC-NKT cells display adjuvant effects for endogenous DCs and NK cells (Kitayama et al. 2016; Yamada et al. 2016; Aoki
et al. 2023)
IL-15 stimulation drives phenotypic maturity of iPSC-NKT cells, with Th1-like phenotype bias, that show tumor cell killing
in vitro (Kitayama et al. 2016) and in vivo (Yamada et al. 2016)
MAIT Preclinical iPSC-MAIT cells exhibit appropriate lineage-specific TCR, appropriate chemokine and cytokine response profiles in vitro, and
population of bone marrow, liver, spleen niches in NSG mice (Wakao et al. 2013)
γδ T Preclinical iPSC-derived γδ T cells can be produced from whole blood PBMC input (Watanabe et al. 2018)
Mimetic CD19CAR γδ T cells, expressing Vγ9Vδ2TCR and high level of NK markers, but no/low phenotypic γδ markers,
show broad in vitro tumor killing (Zeng et al. 2019)
M. Wilson et al.
et al. 2020). Within human patients, FT500 administered in weekly doses, both as
monotherapy and in concert with immune checkpoint blockade, has shown tumor
suppression in patients resistant to blockade therapy alone, with limited adverse
reactions (Hong et al. 2020). Furthermore, more elaborately engineered products
such as FT516, a CAR-NK cell line expressing non-cleavable CD16 for enhanced
antibody-dependent cellular cytotoxicity, has shown similar success for combination
therapy against liquid and solid tumors (Strati et al. 2021). These trials underscore
the preliminary efficacy of such therapies against a broad range of cancers, particu-
larly as a course of therapy for patients resistant to first-line interventions. As clinical
trials continue to establish the efficacy of stem cell-derived lymphocytes, the foun-
dation for introducing such therapies into clinical practice draws nearer to becoming
a reality.
Despite the success achieved through the current autologous model of CAR-T cell
therapy, its clinical utility is limited by its lack of efficacy against solid tumors,
limited scalability for multiple patients, and financial burden placed on cancer
patients. The development of an “off-the-shelf” therapy will reduce the weight of
these expenses by circumventing the need to develop a tailored immunotherapy for
each patient receiving therapy. While other immune cell subtypes, including NK
cells and innate-like T cells, avoid onset of GvHD on account of their intrinsic
recognition mechanisms, necessary steps in isolation, expansion, and genetic engi-
neering present a challenge to maximizing cell product yield. Especially for innate-
like T cells, which represent an extremely low proportion of lymphocytes in
peripheral blood, producing a sufficient number of cells for multiple doses precludes
direct harvest as a cost-effective method.
These barriers to development can be overcome through stem-cell engineering,
which provides a highly scalable and malleable platform that can produce cell
products with limited heterogeneity. The scalability of NK, iNKT, MAIT, and γδ
T cell-based therapies derive from their MHC-independent recognition of tumor
cells, abrogating the onset of GvHD. Among potential sources for stem cell engi-
neering, currently the greatest interest lies in iPSCs due to ease of collection of more
common, terminally differentiated cells for reprogramming. Selecting a lineage
closely related to the desired iPSC-derived lymphocyte can help mitigate functional
variability arising from epigenetic modifications associated with the origin cell type
for iPSCs (Polo et al. 2010; Zhou et al. 2022). Additional variation may arise from
differences in manufacturing protocol, even among products of the same cell
subtype. Additionally, it is necessary to further explore the mechanisms underlying
stem-cell-based therapies. It remains unclear whether there are any discrepancies
between the maturity and cytotoxic capacity of stem cell-derived products versus
their endogenous counterparts. The extensive stem cell manufacturing and engineer-
ing process itself may also lend to differences in therapeutic potency. It is also yet to
be determined if there are any long-term implications associated with the adminis-
tration of stem cell-derived products to patients.
Stem Cell-Derived Cell Therapy for Cancer
8 Discussion
Stem cells, including MSCs, HSCs, ESCs, and iPSCs, represent compelling
candidates for cancer immunotherapy. The current body of research and early-stage
clinical trials provide substantiated evidence supporting the feasibility of engineered
stem cells as a means to generate potent effector cells and bolster antitumor immune
responses for the eradication of cancer. Stem cell engineering and differentiation
confer not only an inexhaustible supply of therapeutic cellular products but also
establish a versatile and adaptable platform for further modifications aimed at enhanc-
ing the antitumor efficacy of resultant cellular products.
The realization of whether stem cell-engineered approaches manifest additional
efficacy in comparison to those employing mature human periphery blood mononu-
clear cell (PBMC)-derived immune cells as therapeutic targets hinges upon clinical
trials. It is conceivable that the two approaches may, in some instances, complement
each other. Ongoing development of clinical trials is poised to provide preliminary
insights.
Given the intricate nature of diseases like cancer, it is probable that the attainment
of enduring therapeutic benefits will necessitate combination therapies. Engineered
stem cell products, characterized by their “off-the-shelf” availability, hold promise
as constituents of treatment regimens that encompass established cancer therapies,
such as surgery, radiation, chemotherapy, and targeted interventions, as well as
emerging cancer immunotherapies, including immune checkpoint inhibitors,
oncolytic viruses, cancer vaccines, and adoptive cellular therapies (Depil et al.
2020; Li et al. 2021a). The adaptability of engineered stem cell products, their
suitability for customization, and scalability to cater to specific cancer types render
them a promising addition to the oncological arsenal, poised to enhance treatment
outcomes and ameliorate patients’ quality of life (Li et al. 2023). As the field
continues to evolve, a methodical and prudent approach is imperative to ensure the
safe and efficient translation of stem cell-based therapies into clinical settings.
Acknowledgments We thank Dr. Lili Yang from the University of California, Los Angeles
(UCLA) for her insightful discussion. Y.-R.L. is supported by a UCLA MIMG M. John Pickett
Post-Doctoral Fellow Award, and a CIRM-BSCRC Postdoctoral Fellowship.
References
Abel AM, Yang C, Thakar MS, Malarkannan S (2018) Natural killer cells: development, matura-
tion, and clinical utilization. Front Immunol 9:1869
Agarwal S, Aznar MA, Rech AJ et al (2023) Deletion of the inhibitory co-receptor CTLA4
enhances and invigorates chimeric antigen receptor T cells. Immunity S1074-7613(23):
00407–00407. https://doi.org/10.1016/j.immuni.2023.09.001
Allen AG, Pattali R, Izzo KM et al (2021) A bicistronic vector expressing CD16 and a membrane
bound IL-15 construct in iPSC derived NK cells increased cytotoxicity and persistence. Blood
138:4809. https://doi.org/10.1182/blood-2021-153258
Aoki T, Motohashi S, Koseki H (2023) Regeneration of invariant natural killer T (iNKT) cells:
application of iPSC technology for iNKT cell-targeted tumor immunotherapy. Inflamm Regen
43:27. https://doi.org/10.1186/s41232-023-00275-5
Awasthi R, Maier HJ, Zhang J, Lim S (2023) Kymriah® (tisagenlecleucel)—an overview of the
clinical development journey of the first approved CAR-T therapy. Hum Vaccines Immunother
19:2210046. https://doi.org/10.1080/21645515.2023.2210046
Baek S, Kim C-S, Kim S-B et al (2011) Combination therapy of renal cell carcinoma or breast
cancer patients with dendritic cell vaccine and IL-2: results from a phase I/II trial. J Transl Med
9:178. https://doi.org/10.1186/1479-5876-9-178
Bagheri Y, Barati A, Aghebati-Maleki A et al (2021) Current progress in cancer immunotherapy
based on natural killer cells. Cell Biol Int 45:2–17. https://doi.org/10.1002/cbin.11465
Beane JD, Lee G, Zheng Z et al (2015) Clinical scale zinc finger nuclease-mediated gene editing of
PD-1 in tumor infiltrating lymphocytes for the treatment of metastatic melanoma. Mol Ther J
Am Soc Gene Ther 23:1380–1390. https://doi.org/10.1038/mt.2015.71
Beckman EM, Porcelli SA, Morita CT et al (1994) Recognition of a lipid antigen by CD1-restricted
αβ+ T cells. Nature 372:691–694. https://doi.org/10.1038/372691a0
Becknell B, Caligiuri MA (2005) Interleukin-2, interleukin-15, and their roles in human natural
killer cells. Adv Immunol 86:209–239. https://doi.org/10.1016/S0065-2776(04)86006-1
Bedford P, Jy J, Collins L, Keizer S (2018) Considering cell therapy product “good manufacturing
practice” status. Front Med 5:118. https://doi.org/10.3389/fmed.2018.00118
Benmebarek M-R, Karches CH, Cadilha BL et al (2019) Killing mechanisms of chimeric antigen
receptor (CAR) T cells. Int J Mol Sci 20:1283. https://doi.org/10.3390/ijms20061283
Blache U, Popp G, Dünkel A et al (2022) Potential solutions for manufacture of CAR T cells in
cancer immunotherapy. Nat Commun 13:5225. https://doi.org/10.1038/s41467-022-32866-0
Bodden M, Häcker A, Röder J et al (2023) Co-expression of an IL-15 superagonist facilitates self-
enrichment of GD2-Targeted CAR-NK cells and mediates potent cell killing in the absence of
IL-2. Cancer 15:4310. https://doi.org/10.3390/cancers15174310
Boti MA, Athanasopoulou K, Adamopoulos PG et al (2023) Recent advances in genome-
engineering strategies. Genes 14:129. https://doi.org/10.3390/genes14010129
Cai S, Hou J, Fujino M et al (2017) iPSC-derived regulatory dendritic cells inhibit allograft rejection
by generating alloantigen-specific regulatory T cells. Stem Cell Rep 8:1174–1189. https://doi.
org/10.1016/j.stemcr.2017.03.020
Cany J, van der Waart AB, Spanholtz J et al (2015) Combined IL-15 and IL-12 drives the
generation of CD34+-derived natural killer cells with superior maturation and alloreactivity
potential following adoptive transfer. Onco Targets Ther 4:e1017701. https://doi.org/10.1080/
2162402X.2015.1017701
Chang M-C, Lee C-N, Chen Y-L et al (2012) Cord blood stem-cell-derived dendritic cells generate
potent antigen-specific immune responses and anti-tumour effects. Clin Sci Lond Engl 1979
123:347–360. https://doi.org/10.1042/CS20110272
Chang Y, Jin G, Luo W et al (2023) Engineered human pluripotent stem cell-derived natural killer
cells with PD-L1 responsive immunological memory for enhanced immunotherapeutic efficacy.
Bioact Mater 27:168–180. https://doi.org/10.1016/j.bioactmat.2023.03.018
Stem Cell-Derived Cell Therapy for Cancer
Chen Y, Zhu X, Liu H et al (2023) The application of HER2 and CD47 CAR-macrophage in
ovarian cancer. J Transl Med 21:654. https://doi.org/10.1186/s12967-023-04479-8
Christodoulou I, Koldobskiy M, Ho WJ et al (2021) Engineered interleukin-15 autocrine signaling
invigorates anti-CD123 CAR-NK cells. Blood 138:2806. https://doi.org/10.1182/blood-
2021-146609
Cichocki F, Bjordahl R, Gaidarova S et al (2020) iPSC-derived NK cells maintain high cytotoxicity
and enhance in vivo tumor control in concert with T cells and anti–PD-1 therapy. Sci Transl
Med 12:eaaz5618. https://doi.org/10.1126/scitranslmed.aaz5618
D’Agostino M, Raje N (2020) Anti-BCMA CAR T-cell therapy in multiple myeloma: can we do
better? Leukemia 34:21–34. https://doi.org/10.1038/s41375-019-0669-4
Datta J, Dai X, Bianchi A et al (2022) Combined MEK and STAT3 inhibition uncovers stromal
plasticity by enriching for cancer-associated fibroblasts with mesenchymal stem cell-like
features to overcome immunotherapy resistance in pancreatic cancer. Gastroenterology 163:
1593–1612. https://doi.org/10.1053/j.gastro.2022.07.076
Depil S, Duchateau P, Grupp SA et al (2020) “Off-the-shelf” allogeneic CAR T cells: development
and challenges. Nat Rev Drug Discov 19:185–199. https://doi.org/10.1038/s41573-019-0051-2
Dhaliwal A, Ravi S (2023) Myelodysplastic syndrome after anti-CD19 chimeric antigen receptor
T-cell therapy: a case series. Cureus 15:e44677. https://doi.org/10.7759/cureus.44677
Dias J, Boulouis C, Gorin J-B et al (2018) The CD4-CD8- MAIT cell subpopulation is a
functionally distinct subset developmentally related to the main CD8+ MAIT cell pool. Proc
Natl Acad Sci 115:E11513–E11522. https://doi.org/10.1073/pnas.1812273115
Ding Q, Regan SN, Xia Y et al (2013) Enhanced efficiency of human pluripotent stem cell genome
editing through replacing TALENs with CRISPRs. Cell Stem Cell 12:393–394. https://doi.org/
10.1016/j.stem.2013.03.006
Ding J, Guyette S, Schrand B et al (2023) Mesothelin-targeting T cells bearing a novel T cell
receptor fusion construct (TRuC) exhibit potent antitumor efficacy against solid tumors. Onco
Targets Ther 12:2182058. https://doi.org/10.1080/2162402X.2023.2182058
Dogan M, Karhan E, Kozhaya L et al (2022) Engineering human MAIT cells with chimeric antigen
receptors for cancer immunotherapy. J Immunol 209:1523–1531. https://doi.org/10.4049/
jimmunol.2100856
Dötsch S, Svec M, Schober K et al (2023) Long-term persistence and functionality of adoptively
transferred antigen-specific T cells with genetically ablated PD-1 expression. Proc Natl Acad Sci
U S A 120:e2200626120. https://doi.org/10.1073/pnas.2200626120
Duan Z, Li Z, Wang Z et al (2023) Chimeric antigen receptor macrophages activated through TLR4
or IFN-γ receptors suppress breast cancer growth by targeting VEGFR2. Cancer Immunol
Immunother 72:3243–3257. https://doi.org/10.1007/s00262-023-03490-8
Eshhar Z, Waks T, Gross G, Schindler DG (1993) Specific activation and targeting of cytotoxic
lymphocytes through chimeric single chains consisting of antibody-binding domains and the
gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci 90:
720–724. https://doi.org/10.1073/pnas.90.2.720
Euchner J, Sprissler J, Cathomen T et al (2021) Natural killer cells generated from human induced
pluripotent stem cells mature to CD56(bright)CD16(+)NKp80(+/-) in-vitro and express
KIR2DL2/DL3 and KIR3DL1. Front Immunol 12:640672. https://doi.org/10.3389/fimmu.
2021.640672
Fang F, Xiao W, Tian Z (2017) NK cell-based immunotherapy for cancer. Semin Immunol 31:37–
54. https://doi.org/10.1016/j.smim.2017.07.009
Fang L, Yuan S, Wang M et al (2023) Recombinant oncolytic adenovirus armed with CCL5, IL-12,
and IFN-γ promotes CAR-T infiltration and proliferation in vivo to eradicate local and distal
tumors. Cell Death Dis 9:328. https://doi.org/10.1038/s41420-023-01626-4
Frank C, Ryan B, Svetlana G et al (2020) iPSC-derived NK cells maintain high cytotoxicity and
enhance in vivo tumor control in concert with T cells and anti–PD-1 therapy. Sci Transl Med 12:
eaaz5618. https://doi.org/10.1126/scitranslmed.aaz5618
M. Wilson et al.
Kagoya Y, Guo T, Yeung B et al (2020) Genetic ablation of HLA class I, class II, and the T-cell
receptor enables allogeneic T cells to be used for adoptive T-cell therapy. Cancer Immunol Res
8:926–936. https://doi.org/10.1158/2326-6066.CIR-18-0508
Kitayama S, Zhang R, Liu T-Y et al (2016) Cellular adjuvant properties, direct cytotoxicity of
re-differentiated Vα24 invariant NKT-like cells from human induced pluripotent stem cells.
Stem Cell Rep 6:213–227. https://doi.org/10.1016/j.stemcr.2016.01.005
Klaihmon P, Kang X, Issaragrisil S, Luanpitpong S (2023) Generation and functional characteriza-
tion of anti-CD19 chimeric antigen receptor-natural killer cells from human induced pluripotent
stem cells. Int J Mol Sci 24:10508. https://doi.org/10.3390/ijms241310508
Klichinsky M, Ruella M, Shestova O et al (2020) Human chimeric antigen receptor macrophages
for cancer immunotherapy. Nat Biotechnol 38:947–953. https://doi.org/10.1038/s41587-020-
0462-y
Lamothe RC, Storlie MD, Espinosa DA et al (2023) Novel CRISPR-associated gene-editing
systems discovered in metagenomic samples enable efficient and specific genome engineering.
CRISPR J 6:243–260. https://doi.org/10.1089/crispr.2022.0089
Lanigan TM, Kopera HC, Saunders TL (2020) Principles of genetic engineering. Genes 11:291.
https://doi.org/10.3390/genes11030291
Li Y, Hermanson DL, Moriarity BS, Kaufman DS (2018) Human iPSC-derived natural killer cells
engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell 23:181–
192.e5. https://doi.org/10.1016/j.stem.2018.06.002
Li Y-R, Dunn ZS, Zhou Y et al (2021a) Development of stem cell-derived immune cells for off-the-
shelf cancer immunotherapies. Cell 10. https://doi.org/10.3390/cells10123497
Li Y-R, Zhou Y, Kim YJ et al (2021b) Development of allogeneic HSC-engineered iNKT cells for
off-the-shelf cancer immunotherapy. Cell Rep Med 2:100449. https://doi.org/10.1016/j.xcrm.
2021.100449
Li Y-R, Zhou Y, Kramer A, Yang L (2021c) Engineering stem cells for cancer immunotherapy.
Trends Cancer. https://doi.org/10.1016/j.trecan.2021.08.004
Li Y-R, Dunn ZS, Garcia G et al (2022a) Development of off-the-shelf hematopoietic stem cell-
engineered invariant natural killer T cells for COVID-19 therapeutic intervention. Stem Cell Res
Ther 13:112. https://doi.org/10.1186/s13287-022-02787-2
Li Y-R, Zeng S, Dunn ZS et al (2022b) Off-the-shelf third-party HSC-engineered iNKT cells for
ameliorating GvHD while preserving GvL effect in the treatment of blood cancers. iScience 25:
104859. https://doi.org/10.1016/j.isci.2022.104859
Li Y-R, Dunn ZS, Yu Y et al (2023) Advancing cell-based cancer immunotherapy through stem cell
engineering. Cell Stem Cell. https://doi.org/10.1016/j.stem.2023.02.009
Liu X, Zhao Y (2018) CRISPR/Cas9 genome editing: fueling the revolution in cancer immunother-
apy. Curr Res Transl Med 66:39–42. https://doi.org/10.1016/j.retram.2018.04.003
Liu E, Tong Y, Dotti G et al (2018) Cord blood NK cells engineered to express IL-15 and a CD19-
targeted CAR show long-term persistence and potent antitumor activity. Leukemia 32:520–531.
https://doi.org/10.1038/leu.2017.226
Lowin B, Hahne M, Mattmann C, Tschopp J (1994) Cytolytic T-cell cytotoxicity is mediated
through perforin and Fas lytic pathways. Nature 370:650–652. https://doi.org/10.1038/
370650a0
Makkouk A, Yang XC, Barca T et al (2021) Off-the-shelf Vδ1 gamma delta T cells engineered with
glypican-3 (GPC-3)-specific chimeric antigen receptor (CAR) and soluble IL-15 display robust
antitumor efficacy against hepatocellular carcinoma. J Immunother Cancer 9:e003441. https://
doi.org/10.1136/jitc-2021-003441
Mehta RS, Rezvani K (2018) Chimeric antigen receptor expressing natural killer cells for the
immunotherapy of cancer. Front Immunol 9
Mishra PJ, Mishra PJ, Glod JW, Banerjee D (2009) Mesenchymal stem cells: flip side of the coin.
Cancer Res 69:1255–1258. https://doi.org/10.1158/0008-5472.CAN-08-3562
M. Wilson et al.
Yeku OO, Brentjens RJ (2016) Armored CAR T-cells: utilizing cytokines and pro-inflammatory
ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans 44:412–418. https://
doi.org/10.1042/BST20150291
Zannikou M, Duffy JT, Levine RN et al (2023) IL15 modification enables CAR T cells to act as a
dual targeting agent against tumor cells and myeloid-derived suppressor cells in GBM.
J Immunother Cancer 11:e006239. https://doi.org/10.1136/jitc-2022-006239
Zeng J, Shahbazi M, Wu C et al (2012) Enhancing immunostimulatory function of human
embryonic stem cell-derived dendritic cells by CD1d overexpression. J Immunol Baltim Md
1950 188:4297–4304. https://doi.org/10.4049/jimmunol.1102343
Zeng J, Tang SY, Wang S (2019) Derivation of mimetic γδ T cells endowed with cancer recognition
receptors from reprogrammed γδ T cell. PLoS One 14:e0216815. https://doi.org/10.1371/
journal.pone.0216815
Zhang Y, Zhang X, Cheng C et al (2017) CRISPR-Cas9 mediated LAG-3 disruption in CAR-T
cells. Front Med 11:554–562. https://doi.org/10.1007/s11684-017-0543-6
Zhang L, Tian L, Dai X et al (2020) Pluripotent stem cell-derived CAR-macrophage cells with
antigen-dependent anti-cancer cell functions. J Hematol Oncol 13:153. https://doi.org/10.1186/
s13045-020-00983-2
Zhang Q, Yang J, Manoharan ENEA et al (2022a) CRISPR/Cas9-mediated gene knockout followed
by negative selection leads to a complete TCR depletion in ortho CAR19 T cells. Bio-Protocol
12:e4485. https://doi.org/10.21769/BioProtoc.4485
Zhang Y, Liu Z, Wei W, Li Y (2022b) TCR engineered T cells for solid tumor immunotherapy. Exp
Hematol Oncol 11:38. https://doi.org/10.1186/s40164-022-00291-0
Zhou Y, Li M, Zhou K et al (2022) Engineering induced pluripotent stem cells for cancer
immunotherapy. Cancer 14:2266. https://doi.org/10.3390/cancers14092266
Zhu Y, Smith DJ, Zhou Y et al (2019) Development of hematopoietic stem cell-engineered
invariant natural killer T cell therapy for cancer. Cell Stem Cell 25:542–557.e9. https://doi.
org/10.1016/j.stem.2019.08.004
Zhu H, Blum RH, Bjordahl R et al (2020a) Pluripotent stem cell–derived NK cells with high-affinity
noncleavable CD16a mediate improved antitumor activity. Blood 135:399–410. https://doi.org/
10.1182/blood.2019000621
Zhu H, You Y, Shen Z, Shi L (2020b) EGFRvIII-CAR-T cells with PD-1 knockout have improved
anti-glioma activity. Pathol Oncol Res 26:2135–2141. https://doi.org/10.1007/s12253-019-
00759-1
Zhu Y, Zuo D, Wang K et al (2023) Mesothelin-targeted CAR-T therapy combined with irinotecan
for the treatment of solid cancer. J Cancer Res Clin Oncol. https://doi.org/10.1007/s00432-023-
05279-9