Adoptive cell therapy (ACT) for cancer shows tremendous potential; however, several challenges preclude its widespread use. These include poor T-cell function in hostile tumor microenvironments, a lack of tumor-specific target antigens, and the high cost and poor scalability of cell therapy manufacturing. Creative genome-editing strategies are beginning to emerge to address each of these limitations, which has initiated the next generation of cell therapy products now entering clinical trials. CRISPR is at the forefront of this revolution, offering a simple and versatile platform for genetic engineering. This review provides a comprehensive overview of CRISPR applications that have advanced ACT.

Significance:

The clinical impact of ACT for cancer can be expanded by implementing specific genetic modifications that enhance the potency, safety, and scalability of cellular products. Here we provide a detailed description of such genetic modifications, highlighting avenues to enhance the therapeutic efficacy and accessibility of ACT for cancer. Furthermore, we review high-throughput CRISPR genetic screens that have unveiled novel targets for cell therapy enhancement.

The promise of adoptive cell therapy (ACT) for cancer has long been recognized, with clinical trials in the 1980s providing some of the first evidence that administration of tumor-reactive immune cells can produce profound tumor regression in some patients (1–4). Building upon this groundwork, Rosenberg and colleagues pioneered the development of ACT using T cells that were isolated directly from the tumor site [tumor-infiltrating lymphocytes (TIL)]. Their method of TIL ACT yielded robust response rates of 40% to 60% in patients with metastatic melanoma (5), a result that has now been replicated across several centers (6–9). More recently, emerging data have demonstrated the efficacy of TIL ACT for other solid tumors, including cervical and head and neck cancers (10, 11).

In the 1990s and early 2000s, the field of ACT moved toward generating T-cell therapies with defined and highly specific tumor reactivities, contrary to the polyclonal and often undefined specificities of TILs. Three dominant approaches emerged that involved (i) isolating and expanding endogenous T cells with defined antigen specificities, (ii) redirecting T-cell specificity via introduction of transgenic T-cell receptors (tgTCR), and (iii) engineering novel T-cell specificities via transduction of chimeric antigen receptors (CAR).

Yee and colleagues demonstrated that high-avidity, tumor-specific T cells can be isolated and expanded from the peripheral blood of patients with cancer (12), and initial clinical use of such cells for ACT elicited antigen-specific tumor regression in patients with melanoma (13). Alternatively, bulk peripheral blood lymphocytes can be genetically engineered to express a tgTCR that recognizes a tumor antigen. Rosenberg and colleagues were the first to demonstrate tumor regression in patients with cancer treated with such TCR-transduced T cells. They reported that 2 of 15 patients with advanced melanoma experienced durable tumor regression following treatment with T cells engineered to recognize a melanocyte differentiation antigen (14). Research is currently ongoing to explore the clinical potential of ACT using TCR-transduced T cells targeting a variety of antigens and cancer types (15).

CAR-engineered T cells have proved to be particularly impactful for the treatment of selected hematologic malignancies. CARs, as originally developed by Eshhar and colleagues (16–18), are fusion proteins that combine the antigen-binding capability of the antibody variable region with T-cell signaling motifs. Thus, CARs are able to recognize cell-surface antigens independent of peptide presentation by major histocompatibility complexes. CAR T cells engineered to recognize the CD19 surface antigen have demonstrated tremendous clinical success for the treatment of CD19+ B-cell malignancies (19–21). This success ultimately led to FDA approval of axicabtagene ciloleucel (Yescarta; package insert available at: https://www.fda.gov/media/108377/download) and tisagenlecleucel (Kymriah; package insert available at: https://www.novartis.us/sites/www.novartis.us/files/kymriah.pdf) in 2017.

Despite these successes, hurdles remain at every step along the pipeline of developing safe, effective cellular therapies for cancer. A dearth of safe target antigens has hampered the development of CAR T cells for broader therapeutic indications beyond CD19+ lymphomas and leukemias. Achieving durable responses of solid tumors to ACT is met by the additional challenges of poor T-cell function in immunosuppressive tumor microenvironments, heterogeneity of antigen expression, and the need for human leukocyte antigen matching in engineered TCR approaches. At the other end of the development pipeline, the high cost and poor scalability of clinical-grade cell therapy manufacturing limits widespread access to these potentially lifesaving treatments.

As genome-editing technologies have become increasingly accessible, their use in cellular therapy has expanded greatly. This has initiated the next generation of cellular products that harbor creative genetic manipulations to improve T cells' potency and safety profiles and that can mitigate immunosuppressive triggers in the tumor microenvironment. Furthermore, genetic engineering approaches that enable use of allogeneic cell sources hold the potential to substantially improve the scalability of cell therapy manufacturing. CRISPR genome-editing techniques are at the forefront of this revolution, offering a versatile platform for applications ranging from multiplexed gene knockout to site-specific transgene insertion to genome-wide genetic screening. The purpose of this review is to detail recent applications of CRISPR that have advanced the field of cellular therapy, beginning with a discussion of specific genetic manipulations that hold the potential to enhance T-cell potency, safety, and scalability and ending with a review of CRISPR screening efforts that have unveiled novel targets for cell therapy enhancement. Emerging applications of CRISPR in non-T cell–based ACTs are also described.

CRISPR: Why and How

CRISPR technology has transformed the landscape of genome editing with its simplicity and versatility (22). Genome editing via CRISPR is achieved using a two-part system comprising a guide RNA (gRNA) and a Cas9 nuclease. The 5′ end of the gRNA directs site specificity via Watson–Crick base pairing, whereas the Cas9 nuclease binds to the 3′ end of the gRNA, positioning it to induce a double-strand break in the target genomic DNA (22). The double-strand break then undergoes repair via the cell's native machinery [nonhomologous end joining and homology-directed repair (HDR)], which often results in small insertions or deletions that disrupt the protein-coding sequence to produce a functional knockout. Furthermore, HDR can be exploited for targeted gene insertion at the CRISPR cut site via inclusion of an HDR template (23).

CRISPR offers a number of notable advantages over earlier genome-editing technologies, such as zinc finger nucleases (ZFN) and transcription activator–like effector nucleases (TALEN). ZFNs and TALENs rely on protein–DNA binding to guide site-specific DNA cleavage. As a result, new ZFN or TALEN proteins must be engineered for each new target site, and design restrictions (particularly for ZFNs) limit the number of sites that can be targeted efficiently (24). ZFNs and TALENs have limited potential for multiplexed genome editing and are not amenable to the generation of large-scale libraries (25). In contrast, the CRISPR system can be designed to target virtually any genomic sequence simply by altering the gRNA sequence. This enables (i) rapid design and implementation of new CRISPR gene knockout/knock-in strategies, (ii) efficient multiplexing via incorporation of multiple gRNAs, and (iii) application of whole-genome CRISPR libraries for high-throughput screening. Finally, the efficiency and specificity of genome editing via CRISPR are generally superior to those of ZFNs and TALENs (25, 26). Specificity is a key safety consideration for clinical applications as off-target mutagenesis poses a risk of oncogenic transformation in genetically engineered cells. Recent technological advances have substantially reduced the risk of off-target mutagenesis with CRISPR, as discussed later in this review. For an in-depth comparison of CRISPR, ZFNs, and TALENs, readers are referred to a review by Khan (25).

Despite the aforementioned strengths of CRISPR genome editing, fully implementing this technology in primary T cells was a challenge for many years. Standard viral transduction approaches to introducing Cas9 and gRNA expression constructs into primary T cells are inefficient (27, 28), likely due to the large size of the Cas9 gene and relative resistance of primary T cells to transduction. Furthermore, stable expression of Cas9 and gRNA following viral integration increases the risk of off-target editing and may result in Cas9 immunogenicity upon adoptive transfer of the edited cells into immunocompetent hosts. DNA nucleofection has been attempted with varying degrees of success but often results in low transfer efficiency and/or high toxicity in T cells, limiting the broad applicability of this method (29–31).

These challenges were overcome with the development of methods to transfect primary T cells with Cas9 ribonucleoprotein (RNP) complexes comprising recombinant Cas9 protein bound to a gRNA (28). Electrotransfer of RNP complexes into primary T cells can be accomplished efficiently without sacrificing cell viability, resulting in highly efficient gene knockout that is observable within about 48 hours. Cas9 delivered via transfection is degraded within 48 to 72 hours, which limits off-target editing and concerns about downstream Cas9 immunogenicity (32). Importantly, applying this technique is feasible within the constraints and timelines of clinical-grade cell therapy manufacturing in compliance with Good Manufacturing Practices, and incorporating a CRISPR gene-editing step is not expected to increase overall production time (33). For the generation for CAR and TCR T-cell therapies, RNP electrotransfer can simply be applied about 1 day prior to standard viral transduction of CAR and TCR transgenes.

Given these benefits, Cas9 RNP electrotransfer has become the primary method of CRISPR editing in T cells and other primary immune cells. This system has proved to be quite versatile, with success seen in (i) single-gene knockout using one or more gRNA sequences (Fig. 1A; refs. 28, 34); (ii) multiplexed gene knockout (Fig. 1B; refs. 35, 36); and (iii) nonviral site-specific gene insertion via inclusion of homologous repair templates in the form of linear DNA oligonucleotides (Fig. 1C; refs. 23, 37, 38). Furthermore, CRISPR editing via Cas9 RNP electrotransfer has been successfully applied to additional primary immune cells, including natural killer (NK) cells (39), B cells (40, 41), and hematopoietic stem and progenitor cells (42–45). Oh and colleagues (46) and Naeimi Kararoudi and colleagues (39) reported detailed protocols describing Cas9 RNP electrotransfer in T and NK cells, respectively.

Figure 1.

Electrotransfer of Cas9 RNP complexes is a versatile and efficient method of CRISPR genome editing in primary T cells. Cas9 RNP complexes comprising Cas9 protein and a gRNA are delivered into activated T cells via electroporation.Genomic editing is detectable about 48 hours after electroporation.A, Multiple RNP complexes targeting the same gene can be delivered simultaneously to increase knockout efficiency. B, Multiple RNP complexes targeting different genes can be delivered simultaneously to achieve multiplexed gene editing. C, RNP complexes can be combined with HDR templates for targeted DNA insertion into the CRISPR cut site.

Figure 1.

Electrotransfer of Cas9 RNP complexes is a versatile and efficient method of CRISPR genome editing in primary T cells. Cas9 RNP complexes comprising Cas9 protein and a gRNA are delivered into activated T cells via electroporation.Genomic editing is detectable about 48 hours after electroporation.A, Multiple RNP complexes targeting the same gene can be delivered simultaneously to increase knockout efficiency. B, Multiple RNP complexes targeting different genes can be delivered simultaneously to achieve multiplexed gene editing. C, RNP complexes can be combined with HDR templates for targeted DNA insertion into the CRISPR cut site.

Close modal

Eliminating the “Brakes” to Create More Potent Cellular Therapies

Resistance of cancer to ACTs can develop through T cell–extrinsic and T cell–intrinsic mechanisms. Extrinsic resistance involves changes in tumor cells that render them resistant to T-cell killing, such as loss of the target antigen (47, 48). Conversely, intrinsic resistance develops when the transferred T cells fail to persist in the patient or when they become hyporesponsive and unable to kill target tumor cells. The latter is particularly relevant in the treatment of solid tumors, where the tumor microenvironment, with its characteristically low pH, lack of nutrients, hypoxia, and abundance of immunosuppressive molecules, presents a major obstacle to T-cell function (49). Furthermore, tumors often upregulate inhibitory proteins such as PD-L1 (CD274) that engage with coinhibitory receptors on the T-cell surface [e.g., PDCD1 (PD-1)] to suppress their antitumor activity (50).

CRISPR can be used to engineer T cells to overcome key pathways of immunosuppression that lead to T cell–intrinsic resistance of cancer to ACTs. The most thoroughly explored example of this is using CRISPR to knock out PD-1, which allows adoptively transferred T cells to resist PD-L1–mediated coinhibition (Fig. 2A; refs. 29–31, 34, 51–57). Preclinical data demonstrate the therapeutic potential of this approach, with improved tumor control reported following treatment with PD-1–knockout T cells in models of gastric cancer (53), leukemia (52, 54), hepatocellular carcinoma (55), triple-negative breast cancer (34), and glioma (31). The first two reports of the use of CRISPR-edited T cells to treat cancer were published in 2020 and involved PD-1 knockout (ClinicalTrials.gov identifiers: NCT03399448 and NCT02793856; Table 1; refs. 29, 35). Although these early clinical studies were not powered to draw conclusions regarding the therapeutic impact of PD-1 knockout, they did provide initial evidence suggesting that CRISPR gene editing is feasible and safe clinically.

Figure 2.

Applications of CRISPR to improve the potency, safety, and scalability of T-cell therapies. A, T-cell therapies are made more potent by knocking out inhibitory receptors (e.g., PD-1) that dampen T-cell function upon ligation with cognate ligands in the tumor microenvironment. B, The safety of TCR T-cell therapies can be improved via CRISPR knockout of the endogenous TCR (eTCR), which prevents formation of novel TCR mismatches with unpredictable neoreactivity. C, With CRISPR gene knockout, it is feasible to generate CAR T-cell therapies from allogeneic T cells. Knockout of the eTCR reduces risk of GvHD whereas knockout of major histocompatibility complex class I (MHC-I) genes prevents rejection of the allogeneic cells upon adoptive transfer. Use of allogeneic cells for CAR T-cell therapy holds the potential to increase the scalability of CAR T-cell manufacturing as many doses can be generated from cells isolated from a single healthy donor. Furthermore, creation of allogeneic T-cell therapies may enable treatment of cancers that are otherwise difficult to treat with autologous T cells. D, CRISPR can be used to knock out target antigens in off-target cells. For example, knocking out CD7 in CD7-directed CAR T cells prevents fratricide of the CAR T cells during manufacturing, enabling CAR T-cell therapy for T cell–based cancers.

Figure 2.

Applications of CRISPR to improve the potency, safety, and scalability of T-cell therapies. A, T-cell therapies are made more potent by knocking out inhibitory receptors (e.g., PD-1) that dampen T-cell function upon ligation with cognate ligands in the tumor microenvironment. B, The safety of TCR T-cell therapies can be improved via CRISPR knockout of the endogenous TCR (eTCR), which prevents formation of novel TCR mismatches with unpredictable neoreactivity. C, With CRISPR gene knockout, it is feasible to generate CAR T-cell therapies from allogeneic T cells. Knockout of the eTCR reduces risk of GvHD whereas knockout of major histocompatibility complex class I (MHC-I) genes prevents rejection of the allogeneic cells upon adoptive transfer. Use of allogeneic cells for CAR T-cell therapy holds the potential to increase the scalability of CAR T-cell manufacturing as many doses can be generated from cells isolated from a single healthy donor. Furthermore, creation of allogeneic T-cell therapies may enable treatment of cancers that are otherwise difficult to treat with autologous T cells. D, CRISPR can be used to knock out target antigens in off-target cells. For example, knocking out CD7 in CD7-directed CAR T cells prevents fratricide of the CAR T cells during manufacturing, enabling CAR T-cell therapy for T cell–based cancers.

Close modal
Table 1.

Ongoing clinical trials involving CRISPR-edited ACT for cancer

CitationClinicalTrials.gov identifierCompany/institutionCRISPR gene targetsCell therapy detailsSummary/status
Trials for which reports are available 
Stadtmauer et al. (35) NCT03399448 University of Pennsylvania PDCD1, TRAC, TRBC NY-ESO-1 TCR T cells Results demonstrate that multiplexed CRISPR/Cas9 genome editing is feasible at clinical scale and suggest acceptable safety 
Xinxin Wang et al.a NCT04264078 Xinqiao Hospital of Chongqing TRAC, CD7 CD7 CAR T cells Results demonstrate deep clinical responses and an acceptable safety profile; GvHD was not observed in any patients treated with allogeneic CAR T cells 
Lu et al. (29) NCT02793856 Sichuan University PDCD1 Peripheral blood lymphocytes Results for patients given CRISPR/Cas9 PD-1–edited T cells suggest that CRISPR editing in adoptively transferred T cells is feasible in a clinical setting and generally safe 
Trials for which reports are not yet available 
 NCT03545815 Chinese PLA General Hospital TCR, PDCD1 Mesothelin CAR T cells Recruiting 
 NCT04035434 CRISPR Therapeutics AG B2M, TRAC CD19 T cells Recruiting 
 NCT03747965 Chinese PLA General Hospital PDCD1 Mesothelin CAR T cells Recruiting 
 NCT03081715 Hangzhou Cancer Hospital PDCD1 Peripheral blood lymphocytes Completed 
 NCT04037566 Xijing Hospital HPK1 CD19 CAR T cells Recruiting 
 NCT03690011 Baylor College of Medicine CD7 CD7 CAR T cells Not yet recruiting 
 NCT04244656 CRISPR Therapeutics AG Unknown B-cell maturation antigen T cells Recruiting 
 NCT03044743 The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School PDCD1 Epstein–Barr virus–specific T cells Recruiting 
 NCT03398967 Chinese PLA General Hospital Unknown CD19 and CD20 or CD22 CAR T cells Recruiting 
 NCT03166878 Chinese PLA General Hospital TCR, B2M CD19 CAR T cells Recruiting 
CitationClinicalTrials.gov identifierCompany/institutionCRISPR gene targetsCell therapy detailsSummary/status
Trials for which reports are available 
Stadtmauer et al. (35) NCT03399448 University of Pennsylvania PDCD1, TRAC, TRBC NY-ESO-1 TCR T cells Results demonstrate that multiplexed CRISPR/Cas9 genome editing is feasible at clinical scale and suggest acceptable safety 
Xinxin Wang et al.a NCT04264078 Xinqiao Hospital of Chongqing TRAC, CD7 CD7 CAR T cells Results demonstrate deep clinical responses and an acceptable safety profile; GvHD was not observed in any patients treated with allogeneic CAR T cells 
Lu et al. (29) NCT02793856 Sichuan University PDCD1 Peripheral blood lymphocytes Results for patients given CRISPR/Cas9 PD-1–edited T cells suggest that CRISPR editing in adoptively transferred T cells is feasible in a clinical setting and generally safe 
Trials for which reports are not yet available 
 NCT03545815 Chinese PLA General Hospital TCR, PDCD1 Mesothelin CAR T cells Recruiting 
 NCT04035434 CRISPR Therapeutics AG B2M, TRAC CD19 T cells Recruiting 
 NCT03747965 Chinese PLA General Hospital PDCD1 Mesothelin CAR T cells Recruiting 
 NCT03081715 Hangzhou Cancer Hospital PDCD1 Peripheral blood lymphocytes Completed 
 NCT04037566 Xijing Hospital HPK1 CD19 CAR T cells Recruiting 
 NCT03690011 Baylor College of Medicine CD7 CD7 CAR T cells Not yet recruiting 
 NCT04244656 CRISPR Therapeutics AG Unknown B-cell maturation antigen T cells Recruiting 
 NCT03044743 The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School PDCD1 Epstein–Barr virus–specific T cells Recruiting 
 NCT03398967 Chinese PLA General Hospital Unknown CD19 and CD20 or CD22 CAR T cells Recruiting 
 NCT03166878 Chinese PLA General Hospital TCR, B2M CD19 CAR T cells Recruiting 

a111th Annual Meeting of the American Association for Cancer Research (abstract available at: https://www.abstractsonline.com/pp8/#!/9045/presentation/10760).

NOTE: Information was gathered through Clinicaltrials.gov unless otherwise noted.

Using CRISPR to knock out PD-1 (or other coinhibitory receptors) in T cells is an alternative to combining ACT with systemic checkpoint-blocking antibodies (e.g., anti–PD-1/PD-L1, anti-CTLA4) and offers attractive benefits. First, checkpoint pathways are engaged throughout the body to promote peripheral tolerance and prevent autoimmunity. Therefore, systemic blockade often results in autoimmune-related side effects (e.g., colitis, pneumonitis; refs. 58, 59). Using CRISPR to eliminate PD-1 specifically in transferred cells limits the risk of breaking peripheral tolerance, especially when any self-reactive TCRs are eliminated from the infusion product (see below). Second, cellular therapies can persist in patients and provide tumor surveillance for many years. CRISPR knockout of a checkpoint receptor protects the transferred cells from PD-1–mediated inhibition for their entire lifetimes and the lifetimes of their progeny. In contrast, checkpoint-blocking antibodies have half-lives of 15 to 25 days and would require prolonged repeat dosing to achieve the same benefit.

Permanent checkpoint elimination via CRISPR knockout is not without limitations. Emerging evidence from studies of patients with cancer treated with PD-1–knockout TCR T cells suggested that cells deficient in PD-1 are less persistent than their PD-1–proficient counterparts, which is consistent with a defect in memory formation (35). In this context, administering a mixed population of T cells may be advantageous, as the PD-1–knockout population may provide potent effector activity in the short term, whereas those with intact PD-1 may persist and provide longer antitumor protection. Alternatively, advanced CRISPR approaches are being explored to provide reversible genetic control of PD-1 expression. In 2019, Qi and colleagues described a system whereby transcriptional repression of PD-1 was transiently induced in CAR T cells upon antigen engagement (abstract presented in SITC Adoptive Cellular Therapies Workshop 2019; https://www.sitcancer.org/education/actworkshop/schedule). This nuanced control of PD-1 may enhance effector function while maintaining memory differentiation and long-term persistence of the edited cells in vivo. Given the complexity of most checkpoint signaling pathways, the multidimensional consequences of CRISPR knockout should be carefully evaluated for each gene target and therapeutic application.

Going beyond the elimination of canonical checkpoint receptors, CRISPR offers the potential to inhibit the activity of metabolic regulators, transcription factors, and signaling molecules that are otherwise challenging to block using conventional antibodies or small molecules. Tang and colleagues (60) used CRISPR to knock out the TGFβ receptor (TGFBR2) in CAR T cells, which rendered these cells unresponsive to high levels of exogenous TGFβ in vitro and improved their antitumor activity in vivo. This is an attractive approach given the pleiotropic roles of TGFβ throughout the body, which present challenges and safety concerns for the development of systemic TGFβ inhibitors (61). Also, Jung and colleagues (62) knocked out the metabolic enzyme diacylglycerol kinase in CAR T cells, which enhanced their effector function and antitumor activity via increased TCR signaling and resistance to immunosuppressive factors. The specificity of CRISPR knockout is also attractive in this context given the roles of diacylglycerol kinases in other cell types and a lack of specific inhibitors targeting this kinase (63).

These methods of enhancing T-cell potency via CRISPR gene editing may ultimately decrease the minimum effective doses of cellular therapies and lessen the burden of manufacturing high T-cell numbers. However, eliminating inherent checks and balances that restrain T-cell responses may exacerbate the risk of cytokine release syndrome (a common and serious side effect of CAR T-cell therapy; ref. 64) and/or cause uncontrolled lymphoproliferation in vivo. These risks should be carefully evaluated for any target in longitudinal preclinical safety studies prior to clinical translation.

Disrupting the Endogenous TCR to Enhance the Safety and Efficacy of T-cell Therapies

Generating engineered TCR T-cell therapies typically involves viral transfer of a tumor-reactive tgTCR into peripheral T cells. Sustained expression of endogenous TCRs in the final T-cell product presents safety and efficacy challenges. First, α and β chains from the endogenous TCR and the tgTCR can form mixed dimers with unpredictable and dangerous neoreactivity (65). Furthermore, mixed dimer formation limits surface expression of the transgenic αβ pair (33). Competition for CD3 binding by the endogenous TCR and tgTCR further limits surface expression of tgTCR complexes (66). Ultimately, reduced tgTCR expression dampens the antigen sensitivity and therapeutic efficacy of the final TCR T-cell product.

CRISPR can be used to efficiently knock out endogenous αβTCR by targeting the genes that encode for the α chain (TRAC) and β chain (TRBC) constant regions (Fig. 2B). Whereas single knockout of TRAC or TRBC is sufficient to increase tgTCR surface expression and antigen sensitivity in some contexts (33, 67), double knockout of both genes is necessary to eliminate the risk of endogenous TCR and tgTCR mispairing and to ensure optimal expression of weakly competitive tgTCRs (33).

Researchers have proposed two methods of endogenous TCR disruption in TCR T cells. First, efficient TRAC/TRBC double knockout can be achieved via single-step transfection with TRAC- and TRBC-specific Cas9 RNP complexes (33, 57). This method is compatible with clinical TCR T-cell manufacturing pipelines, as it can be simply applied about 1 day prior to standard viral transduction of the tgTCR (33). The clinical feasibility of this approach was confirmed in a recent report by Stadtmauer and colleagues (35) describing the use of NY-ESO-1–directed TCR T cells that had undergone TRAC, TRBC, and PDCD1 knockout (ClinicalTrials.gov identifier: NCT03399448; Table 1).

Second, Cas9 RNP complexes can be used to nonvirally guide the integration of tgTCR α- and β-expressing DNA templates into endogenous TRAC and/or TRBC loci (37). These complexes are delivered into cells via electroporation and create double-strand breaks at specific locations within the TRAC and/or TRBC gene. Subsequently, tgTCR templates bearing homology arms (codelivered via electroporation) are integrated into double-strand break sites via HDR. This approach disrupts endogenous TCR expression while putting the transgenes under the control of the endogenous TCR promoters. A strength of this approach is that it does not involve viral transduction for tgTCR integration. Avoiding the lengthy and expensive process of generating clinical-grade viral vectors has the potential to accelerate the development of new TCR T-cell therapies and decrease T-cell therapy production costs. Furthermore, avoiding viral transduction mitigates the risk of oncogenic mutagenesis via random viral integration.

CRISPR has also been used to insert CAR constructs into the TRAC locus (68). Placing the CAR under the transcriptional control of the TRAC gene promotes more consistent and moderate baseline expression of the CAR when compared with random viral integration. This limits tonic signaling at baseline and promotes optimal receptor internalization and recycling upon antigen exposure. Ultimately, these features enhance CAR T-cell potency and delay terminal differentiation and exhaustion of the CAR T cells. An added benefit is that integrating the CAR into the TRAC locus abolishes surface expression of the endogenous TCR. Although disrupting endogenous TCR expression in CAR T cells does not inherently increase CAR T-cell potency, it does limit the risk of undesired autoreactivity and alloreactivity.

Creating Off-the-Shelf Allogeneic Therapies to Broaden Their Disease Applications and Improve Scalability

The majority of TCR and CAR T cells in the clinic today are engineered ex vivo from patients' own (i.e., autologous) T cells. Not only does the use of autologous cells result in costly, time-consuming manufacturing, it also limits the variety of malignancies that can be treated with TCR and CAR T cells. For instance, patients with some of the most aggressive malignancies (e.g., glioblastoma multiforme) often do not have time to wait for an autologous therapy to be manufactured before rapid tumor progression renders them ineligible for treatment (64). For treating T cell–based malignancies, using autologous T cells for CAR or TCR T-cell manufacturing is met with the critical challenge of ensuring that the final T-cell product is not contaminated with malignant T cells that express many of the same surface markers as healthy T cells (69).

Creating off-the-shelf allogeneic cell therapies holds the potential to overcome these challenges. T cells from a single healthy donor could be used to generate many doses of TCR or CAR T-cell products, which would increase the scalability, cost-effectiveness, and standardization of cell therapy manufacturing. Furthermore, manufacturing of allogeneic products could be completed well in advance of patient enrollment, decreasing time to treatment for the patient. However, using allogeneic cells has its own complications, including the risk of life-threatening graft-versus-host disease (GvHD) and alloreactivity resulting in rapid rejection of the transferred cells. Recent advances demonstrate the potential of CRISPR to overcome these challenges and enable the development of allogeneic T-cell therapies that are safe and effective (Fig. 2C).

As described above, allogeneic CAR T cells are particularly attractive for treating T-cell malignancies. Cooper and colleagues (70) developed an off-the-shelf CAR T-cell targeting the CD7 antigen expressed by many T-cell malignancies. The authors rendered their CAR T cells “universal” by using CRISPR to knock out the endogenous TCR, thereby limiting the risk of GvHD. Importantly, they also used CRISPR to knock out CD7 and prevent fratricide of the CAR T cells (see below). A similar approach was recently taken to the clinic by Gracell (ClinicalTrials.gov identifier: NCT04264078; Table 1), with their initial clinical results presented at the 111th Annual Meeting of the American Association for Cancer Research (2020; abstract available at: https://www.abstractsonline.com/pp8/#!/9045/presentation/10760). The authors gave treatment to an initial five patients with T-cell leukemia with allogeneic CD7-directed CAR T cells that had undergone CRISPR knockout of CD7 (to prevent fratricide of the CAR T cells) and TRAC (to prevent GvHD). All five patents experienced complete remission, although one quickly had a relapse, and four had minimal residual disease–negative complete responses at 1 month. Encouragingly, none of these patients experienced acute GvHD. The impact of alloreactivity on the long-term persistence of these CAR T cells remains to be determined.

CRISPR can be used to limit rejection of allogeneic T-cell therapies by knocking out major histocompatibility complex class I expression, which renders the engineered cells invisible to the recipient's CD8+ T cells. Proof-of-principle studies by Liu and colleagues (51) and Choi and colleagues (36) demonstrated the technical feasibility of this approach using CD19- and EGFRvIII-directed CAR T cells, respectively. The authors achieved efficient TCR knockout by targeting the constant region of the TCRα chain (TRAC) and major histocompatibility complex class I knockout by targeting the β2-microglobulin gene (B2M), which encodes for a necessary component of the major histocompatibility complex class I. Whereas major histocompatibility complex class I expression prevents elimination of allogeneic cells by the host's CD8+ T cells, it may lead to downstream activation of NK cells. Additional genetic manipulation, such as via enforced expression of nonclassical human leukocyte antigen molecules (71), could be explored if this challenge comes to fruition.

The initial reports reviewed herein demonstrate the promise of using CRISPR to generate off-the-shelf allogeneic T-cell therapies for cancer. These advances hold the potential to greatly expand the reach of cellular therapies by making production more scalable and cost-effective and by opening doors to the treatment of malignancies such as glioblastoma multiforme and T-cell cancers that are otherwise challenging to treat with autologous products.

Overcoming On-Target Off-Tumor Challenges to Broaden Disease Applications for T-cell Therapies

The power of CAR and TCR T-cell therapy is rooted in the ability of these cells to kill tumors in an antigen-specific manner. However, these T cells cannot distinguish between malignant and heathy cells expressing the same antigen, resulting in so-called on-target off-tumor side effects. This is readily observed in patients given CD19-directed CAR T cells, as tumor regression is associated with long-term B-cell aplasia (72, 73). Nevertheless, patients can tolerate B-cell aplasia, and immunoglobulin replacement therapy can be given to treat hypogammaglobulinemia (72–74). Identifying antigens with similarly acceptable safety profiles has been a major challenge preventing widespread adoption of CAR T-cell therapy for hematologic malignancies.

CRISPR can be used to ameliorate on-target off-tumor challenges by knocking out the target antigen in off-target cells. Candidate acute myeloid leukemia antigens such as CD33 are shared by normal and malignant myeloid cells, so targeting these antigens would result in persistent myeloablation and serious morbidity and/or mortality in the treated patients with leukemia. This can be overcome by combining CD33-directed CAR T-cell therapy with hematopoietic stem cell transplantation using stem cells that have undergone CRISPR deletion of CD33 (43–45). This facilitates eradication of CD33+ acute myeloid leukemia cells with simultaneous reconstitution of the hematopoietic system with CD33-null cells that can function normally while resisting death induced by CAR T cells. This synthetic approach to creating tumor-specific antigens holds the potential to widen the therapeutic window of CAR T-cell therapy and broaden the cancer types for which this therapy is feasible.

Treating T-cell malignancies with CAR T cells is met with an additional on-target off-tumor obstacle: fratricide. Candidate antigens expressed by T-cell tumors, such as CD7 and CD5, are also expressed by CAR T cells themselves, resulting in self-activation and self-killing of CAR T cells during manufacture of the cellular therapy. CRISPR can be used to overcome this challenge by simply knocking out the target antigen in T cells prior to CAR transduction (Fig. 2D). Researchers have successfully applied this approach to generating fratricide-resistant CAR T cells targeting CD7 (70, 75) and CD5 (76), and several such approaches are currently entering clinical trials (Table 1).

Fratricide-resistant CAR T cells can kill not only malignant T cells but also healthy endogenous T cells expressing the same antigens. This makes patients susceptible to life-threatening immunodeficiency via T-cell aplasia. Gomes-Silva and colleagues (75) demonstrated that their fratricide-resistant CD7-directed CAR T cells were responsive to viral peptides in vitro due to the cells' native TCRs. This suggests that autologous CD7-directed CAR T cells can provide patients with T-cell malignancies with some level of protection from pathogens despite elimination of native T cells. On the other hand, Cooper and colleagues (70) argued for the use of allogeneic CAR T cells for the treatment of T-cell malignancies given the risk of malignant contamination of autologous T-cell products and the poor quantity and/or quality of T cells isolated from this heavily pretreated patient population. As discussed above, the use of allogeneic CAR T cells requires TCR knockout to prevent GvHD, which inadvertently eliminates any protection that CAR T cells provide against pathogens via their TCRs.

As an alternative, CAR T-cell therapy for T-cell malignancies can be viewed as a “bridge” to transplantation. Following tumor eradication via treatment with autologous CAR T cells, a patient can receive an allogeneic stem cell transplant, which would terminate CAR T-cell activity (due to alloreactivity/rejection) and reconstitute the hematopoietic system. Alternatively, the target antigen (e.g., CD7) may be knocked out in stem cells (as described previously for CD33; refs. 43–45) to enable long-term coexistence of antitumor CAR T cells and healthy CD7 T cells if the same allogeneic donor is used for CAR T-cell generation and stem cell transplantation.

Applications of CRISPR for ACT Using Cell Types Other Than T Cells

Whereas T cell–based approaches to ACT are by far the most prevalent and mature, the potential of other immune cell subsets in ACT should not be overlooked, and applications of CRISPR in this context are beginning to emerge.

Like T cells, NK cells can directly kill cancer, making them attractive for ACT. NK cells can do so through several mechanisms, including degranulation and antibody-dependent cellular cytotoxicity initiated by receptor [e.g., CD16a (FCGR3A)] binding to the Fc portion of IgG-opsonized target cells (77). In contrast with T cells, NK cell–mediated killing is not antigen-specific, and NK cells do not induce GvHD in the allogeneic setting, making them ideal as off-the-shelf cellular therapy products (77, 78). However, the function of NK cells can be suppressed via exposure to inhibitory factors in the tumor microenvironment and via NK cells' intrinsic checks and balances. Thus, the potency of adoptively transferred NK cells may be enhanced via CRISPR editing.

Pomeroy and colleagues (79) demonstrated that PD-1 knockout increased the functional capacity of primary NK cells in vitro and enhanced their antitumor efficacy in an ovarian cancer model. Furthermore, they demonstrated that CRISPR can be used to enhance antibody-dependent cellular cytotoxicity by preventing CD16a shedding from the NK cell surface via either knockout of ADAM17 (a protease that cleaves CD16a) or CRISPR knock-in of a noncleavable CD16a variant (Fig. 3A; ref. 79). Zhu and colleagues (80) and Daher and colleagues (81) knocked out an important cytokine checkpoint [cytokine-inducible SH2-containing protein (CISH or CIS)] to enhance the metabolic fitness of induced pluripotent stem cell–derived NK cells and IL15-secreting CAR NK cells, respectively. In both reports, CIS knockout improved NK cell effector function in vitro and tumor control upon adoptive transfer in vivo.

Figure 3.

Applications of CRISPR for ACT using cell types other than T cells. A, Killing of target cells by NK cells via antibody-dependent cellular cytotoxicity can be enhanced by CRISPR knockout of ADAM17, a protease that cleaves CD16a from the NK-cell surface. B, Cancer cells often upregulate CD47, which transmits a “don't eat me” signal to macrophages via binding to signal regulatory protein-α (SIRPα) on the macrophage cell surface. Thus, SIRPα knockout in macrophages increases their ability to phagocytose tumor cells.

Figure 3.

Applications of CRISPR for ACT using cell types other than T cells. A, Killing of target cells by NK cells via antibody-dependent cellular cytotoxicity can be enhanced by CRISPR knockout of ADAM17, a protease that cleaves CD16a from the NK-cell surface. B, Cancer cells often upregulate CD47, which transmits a “don't eat me” signal to macrophages via binding to signal regulatory protein-α (SIRPα) on the macrophage cell surface. Thus, SIRPα knockout in macrophages increases their ability to phagocytose tumor cells.

Close modal

Investigators have also applied CRISPR to macrophage-based immunotherapies. Macrophages can engulf cancer cells, and they play a role in tumor control. However, many cancer cells have surface overexpression of CD47, which engages with signal regulatory protein-α (SIRPA) to deliver a “don't eat me” signal to macrophages. Ray and colleagues (82) demonstrated that CRISPR knockout of signal regulatory protein-α in macrophages increased their ability to phagocytose tumor cells in vitro (Fig. 3B). We anticipate that the use of CRISPR to genetically engineer non-T cell–based cellular therapies for cancer will continue to grow in the coming years as these therapies continue to mature.

Emerging Technologies to Enhance the Safety of CRISPR-Engineered Adoptive Cell Therapies

The first two reports of CRISPR-modified T cells being used to treat human cancer were published in 2020 (29, 35), with additional clinical trials now under way (Table 1). As CRISPR-modified cellular therapies continue to advance into clinical use, it is critical to ensure their safety. A primary concern regarding CRISPR-modified ACTs is that induction of double-strand breaks at off-target sites may result in pathogenic cellular modifications and genotoxicity. In the context of multiplexed CRISPR editing, the risk of chromosomal translocations between double-strand break sites presents additional safety concerns. Therefore, prior to clinical use, CRISPR-modified cellular products must be comprehensively evaluated for off-target mutations using an unbiased assay that surveys the entire genome. Chromosomal translocations must also be evaluated when multiplexed editing is performed.

Efforts to limit the off-target effects of CRISPR have yielded exciting technological advances in recent years, holding the potential to enhance the safety profile of CRISPR-modified cellular therapies and ease clinical translation. One particular area of advancement is the generation of high-specificity Cas9 variants characterized by demonstrably lower off-target cleavage compared with wild-type Cas9. These engineered proteins include the so-called high-fidelity Cas9s (spCas9-HF1, ref. 83; HiFi Cas9, ref. 84), enhanced specificity Cas9 [eSpCas9(1.1); ref. 85], evolved Cas9 (evoCas9; ref. 86), and hyperaccurate Cas9 (HypaCas9; ref. 87). The HiFi Cas9 generated by Vakulskas and colleagues is particularly relevant as it retains high on-target editing efficiency when delivered into cells transiently as an RNP complex (84). Furthermore, HiFi Cas9 is now commercially available as a Good Manufacturing Practice–grade protein applicable for clinical cell therapy manufacturing.

Recent innovations in CRISPR-based methodology, such as the development of base editors and prime editors, offer the potential to further increase the safety of CRISPR genome editing by eliminating double-strand break formation altogether. Base editors, which are fusions of a catalytically deficient or nicking Cas9 variant and a deaminase, induce nucleotide modifications within a short DNA window (∼5 base pair) directed by gRNA binding (88, 89). Cytosine (90, 91) and adenine (92) base editors have been developed to induce site-specific C to T or A to G mutations, respectively.

Base editors can be used to (i) alter gene function by introducing a key point mutation (92) or (ii) disrupt gene expression by inducing a premature stop codon (93, 94) or disrupting a splice site (95). A key advantage of this system is that the induced genomic changes occur with single nucleotide–level precision, resulting in an altered genotype that is homogeneous and predictable. This contrasts with the heterogeneous mix of insertions and deletions formed via conventional CRISPR gene knockout. Furthermore, because base editors do not induce double-strand breaks, they are associated with a low risk of off-target insertions and deletions and a low risk of chromosomal translocation in the context of multiplexed editing (95). Encouragingly, Webber and colleagues recently optimized a base editing method for efficient multiplexed gene disruption in primary T cells, demonstrating the power of this technology in the context of ACT (95).

Despite the attractive benefits of CRISPR base editors, this technology is prone to its own characteristic off-target mutations that should be monitored and reduced for any clinical application. Base editors can induce bystander deamination if multiple cytosines or adenines are present within the editing window (88, 91). Furthermore, base editors have been shown to induce single-nucleotide mutations at off-target DNA or even RNA sites (88). Although recent advances in base editor design have reduced these risks (89), off-target mutations should be comprehensively analyzed prior to their clinical use.

CRISPR prime editing offers a novel technique to introduce a wide range of base substitutions, insertions, and deletions without inducing double-strand breaks (96). This technology relies on the fusion of a nicking Cas9 variant and a reverse transcriptase. A modified gRNA directs site-specific editing and serves as a primer for the transcription of a modified DNA sequence at the target site. Prime editing offers a genetic engineering platform that is more versatile than base editing and more efficient than genetic modification via CRISPR-mediated insertion of HDR templates (96). Furthermore, prime editing is characterized by lower off-target modification compared with conventional CRISPR/Cas9 methods (96). Despite the tremendous potential of CRISPR prime editing, further research is required to establish the efficiency of this technique in primary immune cells and to fully characterize its safety with respect to off-target mutations. With further development, we believe that CRISPR base editors and prime editors will allow genetic engineering of adoptive cell therapies with unprecedented precision and flexibility while enhancing safety through the reduction of off-target mutations.

Preclinical Discovery

As detailed in the first half of this review, CRISPR genome editing is a powerful platform for enhancing the efficacy of cellular therapies for cancer. However, determining which genetic alterations among the endless possibilities will be the most therapeutically impactful is difficult. CRISPR-based genetic screening is an unbiased, high-throughput method of testing dozens to tens of thousands of genetic alterations in a single experiment. Genetic screens can be designed in (i) an arrayed format, where individual gRNAs are introduced into cells in the wells of a culture plate or (ii) a pooled format, where barcoded gRNA libraries are combined and simultaneously introduced into single populations of cells. Here we focus on pooled CRISPR screens, as they are generally advantageous for high-throughput analyses, and this technique recently yielded fruitful discoveries for immunotherapy advancement.

In its simplest form, a pooled CRISPR screen involves transducing a cell type of interest with a barcoded library of gRNAs targeting any number of genes (97). The cell line is typically pre-engineered to stably express Cas9, or the Cas9 gene is incorporated into the gRNA library constructs. Following library transduction, edited cells are subjected to a screening assay that involves application of a selective pressure or stimulus (e.g., drug-based treatment, TCR activation) followed by next-generation sequencing to identify enriched and depleted gRNA barcodes in surviving and/or phenotypically isolated cell populations (97). This approach is generally referred to as a loss-of-function screen, as it reveals the consequences of gene loss through CRISPR-mediated knockout. Alternative CRISPR screening techniques rely on CRISPR activation or CRISPR interference to transiently induce or repress gene expression, respectively (97), or CRISPR-mediated gene knock-in to explore the consequences of targeted DNA insertion (23).

Performing pooled CRISPR screens with primary T cells has been a major challenge. Primary T cells are viable in culture for only a short period of time, making the lengthy process of Cas9 transduction, selection, expansion, and subsequent library transduction impractical. Furthermore, T cells are notoriously difficult to transduce, and they do not accept large constructs for simultaneous expression of Cas9 and gRNA.

To circumvent these challenges, researchers performed the first loss-of-function CRISPR screens with T cells using immortalized murine (98) and human (99) cell lines amenable to multiple rounds of transduction and selection (Table 2). They transduced T-cell lines with Cas9 before lentiviral transduction of pooled gRNA libraries spanning the entire genome (for a detailed protocol, see Shang and colleagues, ref. 100). Using this methodology, Okada and colleagues (98) revealed a novel regulatory mechanism of PD-1 expression on the surface of T cells (core fucosylation) that could be targeted pharmacologically to improve the antitumor efficacy of adoptively transferred T cells. Shang and colleagues (99) identified previously unknown regulators of T-cell activation, including FAM49B (CYRIB), enhancing our understanding of TCR signaling and its regulation. Whereas these initial reports demonstrate the potential of genome-wide CRISPR screens with T cells, discoveries made using CRISPR screening with immortalized T-cell lines may not always translate to primary cultures or ACT applications.

Table 2.

Summary of CRISPR genetic screens with T cells

CitationCell typeIn vitro/in vivoScreening detailsScreening assayTechnological advance and/or discovery
Okada et al. (98) Immortalized mouse T-cell line (68–41) In vitro Genome-wide loss-of-function screen to identify genes involved in PD-1 expression induction/maintenance Identified gRNA enriched in sorted PD-1lo cells Discovery: Knocking out genes associated with core fucosylation decreases surface expression of PD-1 on T cells; treating antigen-specific murine T cells with a core fucosylation inhibitor decreases PD-1 expression and improves antigen-specific tumor control (B16-Ovalbumin) in vivo 
Shang et al. (99) Immortalized human T-cell line (Jurkat) In vitro Genome-wide loss-of-function screen to identify genes that regulate T-cell activation gRNA abundance compared in CD69lo and CD69hi cells Discovery: Identified FAM49B as a negative regulator of TCR activation, which is involved in actin cytoskeleton remodeling 
Ting et al. (101) Primary human CD4+ T cells In vitro Proof-of-principle loss-of-function screen with ∼13,000 gRNAs; developed Guide Swap method Proof-of-principle report demonstrated feasibility of this screening methodology Technological advance: Development of Guide Swap to enable large-scale pooled screening with primary human cells (including T cells) that cannot be transduced with Cas9-encoding vectors 
Shifrut et al. (102) Primary human CD8+ T cells In vitro Genome-wide loss-of-function screens to identify genes that regulate T-cell stimulation and suppression; developed SLICE method gRNA abundance compared in CFSEhi and CFSElo T cells after TCR stimulation Technological advance: Development of SLICE screening method to enable large-scale pooled screening in primary human T cells 
     Discovery: Identified genes involved in T-cell suppression that, upon knockout, result in improved antigen-specific killing of tumor cells 
Dong et al. (103) Antigen-specific primary mouse CD8+ T cells In vivo and in vitro Genome-wide loss-of-function screen to identify genes that modulate CD8+ T-cell effector function in vitro and in vivo Identify gRNA associated with increased infiltration in tumors (in vivo); identify gRNA associated with increased T-cell degranulation (in vitroDiscovery: Complementary in vivo and in vitro screening reveals genes that can be targeted to improve T-cell function in the context of immunotherapy; authors validated that Dxh37 knockout in CD8+ T cells improves effector function in vitro and tumor control following adoptive transfer in vivo 
Ye et al. (104) Primary mouse CD8+ T cells In vivo Loss-of-function screen focused on genes encoding for membrane-bound proteins Identify gRNA associated with enhanced T-cell infiltration in glioblastoma tumors following adoptive transfer Discovery: Identified multiple gene targets (novel and known) that, upon knockout, can increase the antitumor efficacy of adoptively transferred T cells; validated the potential of PDIA3 as an immunotherapy target using multiple mouse models with mechanistic characterization 
Wei et al. (105) Antigen-specific primary mouse CD8+ T cells In vivo Loss-of-function screen focused on metabolism-associated genes aimed at identifying factors to improve adoptive cell therapy for cancer Identify gRNA associated with enhanced T-cell infiltration in B16 melanomas following adoptive transfer Discovery: Loss of REGNASE-1 improves CD8+ T-cell accumulation and persistence in the tumor microenvironment, improving antitumor efficacy. Follow-up genome-wide loss-of-function screen identified mechanistic targets of REGNASE-1 and candidates for combination therapy 
Roth et al. (23) Primary human T cells In vitro andin vivo Pooled knock-in screen to identify synthetic constructs that enhance T-cell fitness and antitumor activity; nonviral HDR template library delivered via Cas9 RNP electroporation Identify constructs that enhance T-cell expansion in various contexts (in vitro) and that enhance T-cell infiltration and an antitumor phenotype in tumors (in vivoTechnological advance: Developed methodology to screen large numbers of knock-in constructs in vivo and in vitro using primary T cells Discovery: Identified a novel switch receptor (TGFBR2-41BB) that promotes solid tumor clearance by tgTCR T cells in vivo 
CitationCell typeIn vitro/in vivoScreening detailsScreening assayTechnological advance and/or discovery
Okada et al. (98) Immortalized mouse T-cell line (68–41) In vitro Genome-wide loss-of-function screen to identify genes involved in PD-1 expression induction/maintenance Identified gRNA enriched in sorted PD-1lo cells Discovery: Knocking out genes associated with core fucosylation decreases surface expression of PD-1 on T cells; treating antigen-specific murine T cells with a core fucosylation inhibitor decreases PD-1 expression and improves antigen-specific tumor control (B16-Ovalbumin) in vivo 
Shang et al. (99) Immortalized human T-cell line (Jurkat) In vitro Genome-wide loss-of-function screen to identify genes that regulate T-cell activation gRNA abundance compared in CD69lo and CD69hi cells Discovery: Identified FAM49B as a negative regulator of TCR activation, which is involved in actin cytoskeleton remodeling 
Ting et al. (101) Primary human CD4+ T cells In vitro Proof-of-principle loss-of-function screen with ∼13,000 gRNAs; developed Guide Swap method Proof-of-principle report demonstrated feasibility of this screening methodology Technological advance: Development of Guide Swap to enable large-scale pooled screening with primary human cells (including T cells) that cannot be transduced with Cas9-encoding vectors 
Shifrut et al. (102) Primary human CD8+ T cells In vitro Genome-wide loss-of-function screens to identify genes that regulate T-cell stimulation and suppression; developed SLICE method gRNA abundance compared in CFSEhi and CFSElo T cells after TCR stimulation Technological advance: Development of SLICE screening method to enable large-scale pooled screening in primary human T cells 
     Discovery: Identified genes involved in T-cell suppression that, upon knockout, result in improved antigen-specific killing of tumor cells 
Dong et al. (103) Antigen-specific primary mouse CD8+ T cells In vivo and in vitro Genome-wide loss-of-function screen to identify genes that modulate CD8+ T-cell effector function in vitro and in vivo Identify gRNA associated with increased infiltration in tumors (in vivo); identify gRNA associated with increased T-cell degranulation (in vitroDiscovery: Complementary in vivo and in vitro screening reveals genes that can be targeted to improve T-cell function in the context of immunotherapy; authors validated that Dxh37 knockout in CD8+ T cells improves effector function in vitro and tumor control following adoptive transfer in vivo 
Ye et al. (104) Primary mouse CD8+ T cells In vivo Loss-of-function screen focused on genes encoding for membrane-bound proteins Identify gRNA associated with enhanced T-cell infiltration in glioblastoma tumors following adoptive transfer Discovery: Identified multiple gene targets (novel and known) that, upon knockout, can increase the antitumor efficacy of adoptively transferred T cells; validated the potential of PDIA3 as an immunotherapy target using multiple mouse models with mechanistic characterization 
Wei et al. (105) Antigen-specific primary mouse CD8+ T cells In vivo Loss-of-function screen focused on metabolism-associated genes aimed at identifying factors to improve adoptive cell therapy for cancer Identify gRNA associated with enhanced T-cell infiltration in B16 melanomas following adoptive transfer Discovery: Loss of REGNASE-1 improves CD8+ T-cell accumulation and persistence in the tumor microenvironment, improving antitumor efficacy. Follow-up genome-wide loss-of-function screen identified mechanistic targets of REGNASE-1 and candidates for combination therapy 
Roth et al. (23) Primary human T cells In vitro andin vivo Pooled knock-in screen to identify synthetic constructs that enhance T-cell fitness and antitumor activity; nonviral HDR template library delivered via Cas9 RNP electroporation Identify constructs that enhance T-cell expansion in various contexts (in vitro) and that enhance T-cell infiltration and an antitumor phenotype in tumors (in vivoTechnological advance: Developed methodology to screen large numbers of knock-in constructs in vivo and in vitro using primary T cells Discovery: Identified a novel switch receptor (TGFBR2-41BB) that promotes solid tumor clearance by tgTCR T cells in vivo 

NOTE: Reports are listed in chronological order of publication.

Ting and colleagues (101) and Shifrut and colleagues (102) made breakthroughs in the application of CRISPR screens to primary human T cells by circumventing the need for viral Cas9 transduction (Table 2; refs. 101, 102). Both reports described a method that involves transduction of primary T cells with lentiviral gRNA libraries followed by electroporation-based delivery of Cas9 protein either alone (SLICE method; ref. 102) or complexed with a nontargeting gRNA (Guide Swap method; Fig. 4; ref. 101). In two independent genome-wide screens, Shifrut and colleagues validated the use of this approach to identify regulators of TCR activation and gene deletions that confer resistance of T cells to immunosuppressive adenosine signaling. The techniques developed in these two studies represent a major milestone in the application of CRISPR screening to primary T cells and other immune cells that are difficult to transduce.

Figure 4.

High-throughput CRISPR screening in primary T cells. Novel gene targets to enhance ACT can be identified in an unbiased fashion using pooled CRISPR screens. First, T cells are transduced with a lentiviral library of barcoded gRNA constructs. Second, Cas9 is delivered via electroporation either alone (SLICE method) or in complex with a nontargeting gRNA (Guide Swap method). Finally, the pool of T cells harboring unique CRISPR gene knockouts is subjected to a screening assay, and enriched and/or depleted gRNA barcodes are detected via next-generation sequencing (NGS).

Figure 4.

High-throughput CRISPR screening in primary T cells. Novel gene targets to enhance ACT can be identified in an unbiased fashion using pooled CRISPR screens. First, T cells are transduced with a lentiviral library of barcoded gRNA constructs. Second, Cas9 is delivered via electroporation either alone (SLICE method) or in complex with a nontargeting gRNA (Guide Swap method). Finally, the pool of T cells harboring unique CRISPR gene knockouts is subjected to a screening assay, and enriched and/or depleted gRNA barcodes are detected via next-generation sequencing (NGS).

Close modal

CRISPR screens with human T cells tend to be restricted to in vitro screening assays, in which gRNA enrichment and/or depletion is assessed after application of a stimulus (e.g., TCR stimulation) and/or fluorescence-activated cell sorting of distinct populations (e.g., nonproliferating CFSEhi and proliferating CFSElo cells). Conversely, CRISPR screening with primary murine T cells can be paired with in vivo assays to reveal gene knockouts that promote T-cell infiltration and persistence in complex and immunosuppressive tumor microenvironments. This approach has been used to identify therapeutic targets to improve ACT in models of breast cancer (103), glioblastoma (104), and melanoma (105) as summarized in Table 2.

For CRISPR screening with primary murine T cells, Cas9–knock-in mice are used as sources of T cells that constitutively express Cas9, enabling one-step transduction with a viral library of gRNAs. In the context of cancer immunotherapy, Cas9 mice are typically crossed with a TCR transgenic strain to enable screening with antigen-specific, Cas9-expressing T cells (103, 105). This approach has two limitations. First, genetically crossing mice is a lengthy process that requires a great deal of technical expertise. This adds significant developmental lead time to screening projects and limits the accessibility of this technique. Second, for in vivo screening assays, Cas9+ T cells are typically transferred into wild-type recipient mice that do not express Cas9. Cas9 is an immunogenic bacterial protein whose expression may result in premature clearance of the transferred cells, biasing screening results.

Adapting the aforementioned methodology developed by Ting and colleagues (101) and Shifrut and colleagues (102) to screening of mouse T cells holds the potential to overcome these two limitations. Transient Cas9 delivery via electroporation would prevent the need for crossing TCR transgenic and Cas9–knock-in mice, decreasing project lead time and increasing accessibility of this technique. Furthermore, Cas9 delivered via electroporation would be present in the T cells transiently, reducing the risk of Cas9 immunogenicity upon adoptive transfer.

All T-cell screens described thus far have been loss-of-function screens based on CRISPR gene knockout. Building on this groundwork, more advanced CRISPR-based screening applications are now possible. For example, Roth and colleagues (23) recently reported on the first pooled CRISPR knock-in screen with primary human T cells (Table 2). The authors designed a library of 36 HDR templates encoding for synthetic constructs hypothesized to enhance T-cell fitness. These constructs included dominant-negative receptors, stimulatory switch receptors, transcription factors, and metabolic regulators. The researchers exposed library-modified cells to in vitro and in vivo screening assays to identify the constructs that most reliably increased T-cell fitness and antitumor function. They ultimately identified and validated that a novel switch receptor with the extracellular domain of the TGFβ receptor (TGFBR2) and the intracellular signaling domain of 4-1BB (TNFRSF9) increased T-cell fitness in vitro and enhanced tumor clearance by adoptively transferred T cells in vivo.

The CRISPR screening methods discussed herein are expected to have a substantial positive impact on the development of more potent ACTs. Genome-wide loss-of-function screens will continue to reveal novel targets that can be manipulated to enhance T-cell function and overcome key pathways of immunosuppression. Meanwhile, focused screens of 10 to 50 genetic alterations will enable researchers to systematically and efficiently evaluate the therapeutic benefit of numerous candidate alterations, accelerating the translational development of genetic engineering approaches that are expected to have the most profound clinical impact.

Lentiviral Insertional Mutagenesis as a Genetic Screening Platform Applicable to Patients with Cancer

Patients with cancer are the most relevant subjects for genetic screening assays aimed at uncovering mechanisms to improve the efficacy of cellular immunotherapy in a given patient population. Whereas the aforementioned CRISPR screens cannot be applied to human subjects directly, analyzing the consequences of lentiviral insertional mutagenesis from a screening perspective can reveal gene disruptions that enhance the antitumor efficacy and persistence of cellular therapies in the clinic. This provides a platform for reverse translational research that is expected to yield therapeutic advances with direct relevance to specific patient populations.

Fraietta and colleagues (106) made the serendipitous observation that lentiviral integration of a CD19-directed CAR construct in the TET2 gene was associated with remission in a patient with chronic lymphocytic leukemia. Despite infusing a polyclonal pool of cells harboring unique lentiviral integration sites, more than 90% of the persisting CAR T cells observed during peak response originated from a single clone with TET2 integration. In-depth analysis demonstrated that TET2 disruption was associated with profound CAR T-cell expansion in vivo accompanied by differentiation into a central memory phenotype, prolonged persistence, and ultimately robust antitumor efficacy. This highlights the therapeutic potential of disrupting TET2 (e.g., via CRISPR knockout) to enhance the therapeutic performance of CAR T cells in patients with chronic lymphocytic leukemia.

Building upon this initial observation, Nobles and colleagues (107) set out to correlate lentiviral integration sites with clonal expansion and clinical outcomes in an additional 39 patients with chronic lymphocytic leukemia given treatment with CD19-directed CAR T cells. Indeed, the authors identified a number of genes at integration sites that were associated with enhanced proliferation and long-term persistence of CAR T cells. These genes may prove to be valuable targets for improving the therapeutic efficacy of CAR T cells in patients with chronic lymphocytic leukemia. This powerful approach can be expanded to identify therapeutic targets in additional patient populations already being given virally engineered T-cell therapies. Taking any gene targets identified using this approach back to the laboratory to rigorously test their safety and therapeutic tractability will be important. This approach of reverse translational research holds tremendous potential for the development of therapeutic strategies that have direct clinical relevance.

The development of methods to apply CRISPR genome editing to primary T cells has opened the door to numerous advances aimed at improving the potency, safety, and scalability of ACTs. First, CRISPR knockout of inhibitory factors that restrain T-cell function increases the antitumor efficacy of ACT in preclinical studies. This offers the potential for treating cancer with lower doses of highly potent gene-modified T cells than those doses used currently, which would lessen the burden of manufacturing massive cell numbers for clinical infusion. Second, novel techniques of guiding TCR and CAR transgene integration into the native TCR locus offer an alternative to laborious and costly viral transduction with the added benefit of improving safety via elimination of endogenous TCR expression. Third, CRISPR knockout of the TCRα gene paved the way for the first clinical use of allogeneic CAR T cells with no evidence of acute GvHD. Use of allogeneic T cells for CAR T-cell generation is expected to dramatically reduce the cost of T-cell therapy manufacturing, as cells from a single healthy donor can be used to manufacture many CAR T-cell doses. Furthermore, access to off-the-shelf T-cell products is expected to be particularly impactful for patients with rapidly progressing cancer who do not have time to wait for generation of an autologous therapy. Fourth, creative methods of overcoming on-target off-tumor toxicity offer the potential to extend the success of CAR T-cell therapy to indications beyond CD19+ B-cell malignancies. About a dozen open clinical trials involve CRISPR-edited T-cell therapies, and we anticipate the number to grow continually as the many preclinical approaches described herein continue to mature. Furthermore, recent breakthroughs have resulted in a spike in CRISPR genetic screening with primary T cells. These screens have yielded fruitful discoveries and novel targets for enhancing the efficacy of ACT. Continued use of CRISPR screens will accelerate preclinical discovery and enable systematic prioritization of genetic edits that have the greatest potential therapeutic impact.

S.M. Fix reports grants from National Institutes of Health and Cancer Prevention and Research Institute of Texas during the conduct of the study; in addition, Dr. Fix has a patent for “Engineered T Cells and Tumor-Infiltrating Lymphocytes to Overcome Immunosuppression in the Tumor Microenvironment” pending. A.A. Jazaeri reports clinical trial research support from Iovance, BMS, AstraZeneca, Aravive, Merck, Eli Lilly, and Genentech-Roche and personal fees from AvengeBio and Nuprobe outside the submitted work. P. Hwu reports personal fees from Dragonfly, Immatics, GSK, and Sanofi outside the submitted work; in addition, Dr. Hwu has a patent for “Engineered T Cells and Tumor-Infiltrating Lymphocytes to Overcome Immunosuppression in the Tumor Microenvironment” pending.

The authors thank Rachel Hicklen and Donald Norwood of MD Anderson Scientific Publications, Research Medical Library for literature searching and editing of the manuscript, respectively. The authors thank David Aten of the MD Anderson Department of Creative Communications for figure illustrations. The authors wish to acknowledge generous support from the Dunwoody-Edwards Ovarian Cancer Philanthropic Fund and the Miriam and Jim Mulva Research Fund. S.M. Fix was supported by the CPRIT Research Training Program (RP170067), the Center for Clinical and Translational Sciences TL1 Training Program (NIH Grant No. TL1 TR003169), and an NIH/NCI National Research Service Award (F32CA253968). The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

1.
Rosenberg
SA
,
Lotze
MT
,
Muul
LM
,
Leitman
S
,
Chang
AE
,
Ettinghausen
SE
, et al
Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer
.
N Engl J Med
1985
;
313
:
1485
92
.
2.
Thompson
JA
,
Shulman
KL
,
Benyunes
MC
,
Lindgren
CG
,
Collins
C
,
Lange
PH
, et al
Prolonged continuous intravenous infusion interleukin-2 and lymphokine-activated killer-cell therapy for metastatic renal cell carcinoma
.
J Clin Oncol
1992
;
10
:
960
8
.
3.
Rosenberg
SA
,
Lotze
MT
,
Muul
LM
,
Chang
AE
,
Avis
FP
,
Leitman
S
, et al
A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone
.
N Engl J Med
1987
;
316
:
889
97
.
4.
Thompson
JA
,
Lee
DJ
,
Lindgren
CG
,
Benz
LA
,
Collins
C
,
Shuman
WP
, et al
Influence of schedule of interleukin 2 administration on therapy with interleukin 2 and lymphokine activated killer cells
.
Cancer Res
1989
;
49
:
235
40
.
5.
Rosenberg
SA
,
Packard
BS
,
Aebersold
PM
,
Solomon
D
,
Topalian
SL
,
Toy
ST
, et al
Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report
.
N Engl J Med
1988
;
319
:
1676
80
.
6.
Radvanyi
LG
,
Bernatchez
C
,
Zhang
M
,
Fox
PS
,
Miller
P
,
Chacon
J
, et al
Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients
.
Clin Cancer Res
2012
;
18
:
6758
70
.
7.
Besser
MJ
,
Shapira-Frommer
R
,
Itzhaki
O
,
Treves
AJ
,
Zippel
DB
,
Levy
D
, et al
Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies
.
Clin Cancer Res
2013
;
19
:
4792
800
.
8.
Andersen
R
,
Donia
M
,
Ellebaek
E
,
Borch
TH
,
Kongsted
P
,
Iversen
TZ
, et al
Long-lasting complete responses in patients with metastatic melanoma after adoptive cell therapy with tumor-infiltrating lymphocytes and an attenuated IL2 regimen
.
Clin Cancer Res
2016
;
22
:
3734
45
.
9.
Goff
SL
,
Dudley
ME
,
Citrin
DE
,
Somerville
RP
,
Wunderlich
JR
,
Danforth
DN
, et al
Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma
.
J Clin Oncol
2016
;
34
:
2389
97
.
10.
Jazaeri
AA
,
Zsiros
E
,
Amaria
RN
,
Artz
AS
,
Edwards
RP
,
Wenham
RM
, et al
Safety and efficacy of adoptive cell transfer using autologous tumor infiltrating lymphocytes (LN-145) for treatment of recurrent, metastatic, or persistent cervical carcinoma
.
J Clin Oncol
2019
;
37
:
2538
.
11.
Chesney
JA
,
Lutzky
J
,
Thomas
SS
,
Nieva
JJ
,
Munoz Couselo
E
,
Martin-Liberal
J
, et al
A phase II study of autologous tumor infiltrating lymphocytes (TIL, LN-144/LN-145) in patients with solid tumors
.
J Clin Oncol
2019
;
37
:
TPS2648
.
12.
Yee
C
,
Savage
PA
,
Lee
PP
,
Davis
MM
,
Greenberg
PD
. 
Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers
.
J Immunol
1999
;
162
:
2227
34
.
13.
Yee
C
,
Thompson
JA
,
Byrd
D
,
Riddell
SR
,
Roche
P
,
Celis
E
, et al
Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells
.
Proc Natl Acad Sci U S A
2002
;
99
:
16168
73
.
14.
Morgan
RA
,
Dudley
ME
,
Wunderlich
JR
,
Hughes
MS
,
Yang
JC
,
Sherry
RM
, et al
Cancer regression in patients after transfer of genetically engineered lymphocytes
.
Science
2006
;
314
:
126
9
.
15.
Zhang
J
,
Wang
L
. 
The emerging world of TCR-T cell trials against cancer: a systematic review
.
Technol Cancer Res Treat
2019
;
18
:
1533033819831068
16.
Gross
G
,
Waks
T
,
Eshhar
Z
. 
Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity
.
Proc Natl Acad Sci U S A
1989
;
86
:
10024
8
.
17.
Eshhar
Z
,
Waks
T
,
Gross
G
,
Schindler
DG
. 
Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the γ or ζ subunits of the immunoglobulin and T-cell receptors
.
Proc Natl Acad Sci U S A
1993
;
90
:
720
4
.
18.
Hwu
P
,
Shafer
GE
,
Treisman
J
,
Schindler
DG
,
Gross
G
,
Cowherd
R
, et al
Lysis of ovarian cancer cells by human lymphocytes redirected with a chimeric gene composed of an antibody variable region and the Fc receptor γ chain
.
J Exp Med
1993
;
178
:
361
6
.
19.
Kalos
M
,
Levine
BL
,
Porter
DL
,
Katz
S
,
Grupp
SA
,
Bagg
A
, et al
T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia
.
Sci Transl Med
2011
;
3
:
95ra73
.
20.
Kochenderfer
JN
,
Wilson
WH
,
Janik
JE
,
Dudley
ME
,
Stetler-Stevenson
M
,
Feldman
SA
, et al
Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19
.
Blood
2010
;
116
:
4099
102
.
21.
Brentjens
RJ
,
Davila
ML
,
Riviere
I
,
Park
J
,
Wang
X
,
Cowell
LG
, et al
CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia
.
Sci Transl Med
2013
;
5
:
177ra38
.
22.
Doudna
JA
,
Charpentier
E
. 
Genome editing: the new frontier of genome engineering with CRISPR-Cas9
.
Science
2014
;
346
:
1258096
.
23.
Roth
TL
,
Li
PJ
,
Blaeschke
F
,
Nies
JF
,
Apathy
R
,
Mowery
C
, et al
Pooled knockin targeting for genome engineering of cellular immunotherapies
.
Cell
2020
;
181
:
728
44
.
24.
LaFountaine
JS
,
Fathe
K
,
Smyth
HDC
. 
Delivery and therapeutic applications of gene editing technologies ZFNs, TALENs, and CRISPR/Cas9
.
Int J Pharm
2015
;
494
:
180
94
.
25.
Khan
SH
. 
Genome-editing technologies: concept, pros, and cons of various genome-editing techniques and bioethical concerns for clinical application
.
Mol Ther Nucleic Acids
2019
;
16
:
326
34
.
26.
Osborn
MJ
,
Webber
BR
,
Knipping
F
,
Lonetree
CL
,
Tennis
N
,
DeFeo
AP
, et al
Evaluation of TCR gene editing achieved by TALENs, CRISPR/Cas9, and megaTAL nucleases
.
Mol Ther
2016
;
24
:
570
81
.
27.
Wang
W
,
Ye
C
,
Liu
J
,
Zhang
D
,
Kimata
JT
,
Zhou
P
. 
CCR5 gene disruption via lentiviral vectors expressing Cas9 and single guided RNA renders cells resistant to HIV-1 infection
.
PLoS One
2014
;
9
:
e115987
.
28.
Seki
A
,
Rutz
S
. 
Optimized RNP transfection for highly efficient CRI SPR/Cas9-mediated gene knockout in primary T cells
.
J Exp Med
2018
;
215
:
985
97
.
29.
Lu
Y
,
Xue
J
,
Deng
T
,
Zhou
X
,
Yu
K
,
Deng
L
, et al
Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer
.
Nat Med
2020
;
26
:
732
40
.
30.
Su
S
,
Hu
B
,
Shao
J
,
Shen
B
,
Du
J
,
Du
Y
, et al
CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients
.
Sci Rep
2016
;
6
:
20070
.
31.
Hu
B
,
Zou
Y
,
Zhang
L
,
Tang
J
,
Niedermann
G
,
Firat
E
, et al
Nucleofection with plasmid DNA for CRISPR/Cas9-mediated inactivation of programmed cell death protein 1 in CD133-specific CAR T cells
.
Hum Gene Ther
2019
;
30
:
446
58
.
32.
Liang
X
,
Potter
J
,
Kumar
S
,
Zou
Y
,
Quintanilla
R
,
Sridharan
M
, et al
Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection
.
J Biotechnol
2015
;
208
:
44
53
.
33.
Morton
LT
,
Reijmers
RM
,
Wouters
AK
,
Kweekel
C
,
Remst
DFG
,
Pothast
CR
, et al
Simultaneous deletion of endogenous TCRalphabeta for TCR gene therapy creates an improved and safe cellular therapeutic
.
Mol Ther
2020
;
28
:
64
74
.
34.
Hu
W
,
Zi
Z
,
Jin
Y
,
Li
G
,
Shao
K
,
Cai
Q
, et al
CRISPR/Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell effector functions
.
Cancer Immunol Immunother
2019
;
68
:
365
77
.
35.
Stadtmauer
EA
,
Fraietta
JA
,
Davis
MM
,
Cohen
AD
,
Weber
KL
,
Lancaster
E
, et al
CRISPR-engineered T cells in patients with refractory cancer
.
Science
2020
;
367
:
eaba7365
.
36.
Choi
BD
,
Yu
X
,
Castano
AP
,
Darr
H
,
Henderson
DB
,
Bouffard
AA
, et al
CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma
.
J Immunother Cancer
2019
;
7
:
304
.
37.
Roth
TL
,
Puig-Saus
C
,
Yu
R
,
Shifrut
E
,
Carnevale
J
,
Li
PJ
, et al
Reprogramming human T cell function and specificity with non-viral genome targeting
.
Nature
2018
;
559
:
405
9
.
38.
Schumann
K
,
Lin
S
,
Boyer
E
,
Simeonov
DR
,
Subramaniam
M
,
Gate
RE
, et al
Generation of knock-in primary human T cells using Cas9 ribonucleoproteins
.
Proc Natl Acad Sci U S A
2015
;
112
:
10437
42
.
39.
Naeimi Kararoudi
M
,
Dolatshad
H
,
Trikha
P
,
Hussain
S-RA
,
Elmas
E
,
Foltz
JA
, et al
Generation of knock-out primary and expanded human NK cells using Cas9 ribonucleoproteins
.
J Vis Exp
2018
;
136
:
58237
.
40.
Hung
KL
,
Meitlis
I
,
Hale
M
,
Chen
CY
,
Singh
S
,
Jackson
SW
, et al
Engineering protein-secreting plasma cells by homology-directed repair in primary human B cells
.
Mol Ther
2018
;
26
:
456
67
.
41.
Wu
C-AM
,
Roth
TL
,
Baglaenko
Y
,
Ferri
DM
,
Brauer
P
,
Zuniga-Pflucker
JC
, et al
Genetic engineering in primary human B cells with CRISPR-Cas9 ribonucleoproteins
.
J Immunol Methods
2018
;
457
:
33
40
.
42.
Xu
L
,
Wang
J
,
Liu
Y
,
Xie
L
,
Su
B
,
Mou
D
, et al
CRISPR-edited stem cells in a patient with HIV and acute lymphocytic leukemia
.
N Engl J Med
2019
;
381
:
1240
7
.
43.
Kim
MY
,
Yu
KR
,
Kenderian
SS
,
Ruella
M
,
Chen
S
,
Shin
TH
, et al
Genetic inactivation of CD33 in hematopoietic stem cells to enable CAR T cell immunotherapy for acute myeloid leukemia
.
Cell
2018
;
173
:
1439
53
.
44.
Humbert
O
,
Laszlo
GS
,
Sichel
S
,
Ironside
C
,
Haworth
KG
,
Bates
OM
, et al
Engineering resistance to CD33-targeted immunotherapy in normal hematopoiesis by CRISPR/Cas9-deletion of CD33 exon 2
.
Leukemia
2019
;
33
:
762
808
.
45.
Borot
F
,
Wang
H
,
Ma
Y
,
Jafarov
T
,
Raza
A
,
Ali
AM
, et al
Gene-edited stem cells enable CD33-directed immune therapy for myeloid malignancies
.
Proc Natl Acad Sci U S A
2019
;
116
:
11978
87
.
46.
Oh
SA
,
Seki
A
,
Rutz
S
. 
Ribonucleoprotein transfection for CRISPR/Cas9-mediated gene knockout in primary T cells
.
Curr Protoc Immunol
2019
;
124
:
e69
.
47.
Shah
NN
,
Fry
TJ
. 
Mechanisms of resistance to CAR T cell therapy
.
Nat Rev Clin Oncol
2019
;
16
:
372
85
.
48.
Fry
TJ
,
Shah
NN
,
Orentas
RJ
,
Stetler-Stevenson
M
,
Yuan
CM
,
Ramakrishna
S
, et al
CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy
.
Nat Med
2018
;
24
:
20
8
.
49.
Cheng
J
,
Zhao
L
,
Zhang
Y
,
Qin
Y
,
Guan
Y
,
Zhang
T
, et al
Understanding the mechanisms of resistance to CAR T-cell therapy in malignancies
.
Front Oncol
2019
;
9
:
1237
.
50.
Chen
G
,
Huang
AC
,
Zhang
W
,
Zhang
G
,
Wu
M
,
Xu
W
, et al
Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response
.
Nature
2018
;
560
:
382
6
.
51.
Liu
X
,
Zhang
Y
,
Cheng
C
,
Cheng
AW
,
Zhang
X
,
Li
N
, et al
CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells
.
Cell Res
2017
;
27
:
154
7
.
52.
Ren
J
,
Liu
X
,
Fang
C
,
Jiang
S
,
June
CH
,
Zhao
Y
. 
Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition
.
Clin Cancer Res
2017
;
23
:
2255
66
.
53.
Su
S
,
Zou
Z
,
Chen
F
,
Ding
N
,
Du
J
,
Shao
J
, et al
CRISPR-cas9-mediated disruption of PD-1 on human T cells for adoptive cellular therapies of EBV positive gastric cancer
.
Oncoimmunology
2017
;
6
:
e1249558
.
54.
Rupp
LJ
,
Schumann
K
,
Roybal
KT
,
Gate
RE
,
Ye
CJ
,
Lim
WA
, et al
CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells
.
Sci Rep
2017
;
7
:
737
.
55.
Guo
X
,
Jiang
H
,
Shi
B
,
Zhou
M
,
Zhang
H
,
Shi
Z
, et al
Disruption of PD-1 enhanced the anti-tumor activity of chimeric antigen receptor T cells against hepatocellular carcinoma
.
Front Pharmacol
2018
;
9
:
1118
.
56.
Zhang
C
,
Peng
Y
,
Hublitz
P
,
Zhang
H
,
Dong
T
. 
Genetic abrogation of immune checkpoints in antigen-specific cytotoxic T-lymphocyte as a potential alternative to blockade immunotherapy
.
Sci Rep
2018
;
8
:
5549
.
57.
Ouchi
Y
,
Patil
A
,
Tamura
Y
,
Nishimasu
H
,
Negishi
A
,
Paul
SK
, et al
Generation of tumor antigen-specific murine CD8+ T cells with enhanced anti-tumor activity via highly efficient CRISPR/Cas9 genome editing
.
Int Immunol
2018
;
30
:
141
54
.
58.
Chan
KK
,
Bass
AR
. 
Autoimmune complications of immunotherapy: pathophysiology and management
.
BMJ
2020
;
369
:
m736
.
59.
Xing
P
,
Zhang
F
,
Wang
G
,
Xu
Y
,
Li
C
,
Wang
S
, et al
Incidence rates of immune-related adverse events and their correlation with response in advanced solid tumours treated with NIVO or NIVO + IPI: a systematic review and meta-analysis
.
J Immunother Cancer
2019
;
7
:
341
.
60.
Tang
N
,
Cheng
C
,
Zhang
X
,
Qiao
M
,
Li
N
,
Mu
W
, et al
TGF-beta inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors
.
JCI Insight
2020
;
5
:
e133977
.
61.
Smith
AL
,
Robin
TP
,
Ford
HL
. 
Molecular pathways: targeting the TGF-β pathway for cancer therapy
.
Clin Cancer Res
2012
;
18
:
4514
21
.
62.
Jung
IY
,
Kim
YY
,
Yu
HS
,
Lee
M
,
Kim
S
,
Lee
J
. 
CRISPR/Cas9-mediated knockout of DGK improves antitumor activities of human T cells
.
Cancer Res
2018
;
78
:
4692
703
.
63.
Purow
B
. 
Molecular pathways: targeting diacylglycerol kinase alpha in cancer
.
Clin Cancer Res
2015
;
21
:
5008
12
.
64.
Santomasso
B
,
Bachier
C
,
Westin
J
,
Rezvani
K
,
Shpall
EJ
. 
The other side of CAR T-cell therapy: cytokine release syndrome, neurologic toxicity, and financial burden
.
Am Soc Clin Oncol Educ Book
2019
;
39
:
433
44
.
65.
van Loenen
MM
,
de Boer
R
,
Amir
AL
,
Hagedoorn
RS
,
Volbeda
GL
,
Willemze
R
, et al
Mixed T cell receptor dimers harbor potentially harmful neoreactivity
.
Proc Natl Acad Sci U S A
2010
;
107
:
10972
7
.
66.
Ahmadi
M
,
King
JW
,
Xue
S-A
,
Voisine
C
,
Holler
A
,
Wright
GP
, et al
CD3 limits the efficacy of TCR gene therapy in vivo
.
Blood
2011
;
118
:
3528
37
.
67.
Legut
M
,
Dolton
G
,
Mian
AA
,
Ottmann
OG
,
Sewell
AK
. 
CRISPR-mediated TCR replacement generates superior anticancer transgenic t cells
.
Blood
2018
;
131
:
311
22
.
68.
Eyquem
J
,
Mansilla-Soto
J
,
Giavridis
T
,
Van Der Stegen
SJC
,
Hamieh
M
,
Cunanan
KM
, et al
Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection
.
Nature
2017
;
543
:
113
7
.
69.
Alcantara
M
,
Tesio
M
,
June
CH
,
Houot
R
. 
CAR T-cells for T-cell malignancies: challenges in distinguishing between therapeutic, normal, and neoplastic T-cells
.
Leukemia
2018
;
32
:
2307
15
.
70.
Cooper
ML
,
Choi
J
,
Staser
K
,
Ritchey
JK
,
Devenport
JM
,
Eckardt
K
, et al
An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies
.
Leukemia
2018
;
32
:
1970
83
.
71.
Torikai
H
,
Reik
A
,
Soldner
F
,
Warren
EH
,
Yuen
C
,
Zhou
Y
, et al
Toward eliminating HLA class I expression to generate universal cells from allogeneic donors
.
Blood
2013
;
122
:
1341
9
.
72.
Porter
DL
,
Hwang
W-T
,
Frey
NV
,
Lacey
SF
,
Shaw
PA
,
Loren
AW
, et al
Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia
.
Sci Transl Med
2015
;
7
:
303ra139
.
73.
Maude
SL
,
Laetsch
TW
,
Buechner
J
,
Rives
S
,
Boyer
M
,
Bittencourt
H
, et al
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med
2018
;
378
:
439
48
.
74.
Hill
JA
,
Giralt
S
,
Torgerson
TR
,
Lazarus
HM
. 
CAR-T - and a side order of IgG, to go? – Immunoglobulin replacement in patients receiving CAR-T cell therapy
.
Blood Rev
2019
;
38
:
100596
.
75.
Gomes-Silva
D
,
Srinivasan
M
,
Sharma
S
,
Lee
CM
,
Wagner
DL
,
Davis
TH
, et al
CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies
.
Blood
2017
;
130
:
285
96
.
76.
Raikar
SS
,
Fleischer
LC
,
Moot
R
,
Fedanov
A
,
Paik
NY
,
Knight
KA
, et al
Development of chimeric antigen receptors targeting T-cell malignancies using two structurally different anti-CD5 antigen binding domains in NK and CRISPR-edited T cell lines
.
Oncoimmunology
2018
;
7
:
e1407898
.
77.
Daher
M
,
Rezvani
K
. 
Next generation natural killer cells for cancer immunotherapy: the promise of genetic engineering
.
Curr Opin Immunol
2018
;
51
:
146
53
.
78.
Liu
E
,
Marin
D
,
Banerjee
P
,
Macapinlac
HA
,
Thompson
P
,
Basar
R
, et al
Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors
.
N Engl J Med
2020
;
382
:
545
53
.
79.
Pomeroy
EJ
,
Hunzeker
JT
,
Kluesner
MG
,
Lahr
WS
,
Smeester
BA
,
Crosby
MR
, et al
A genetically engineered primary human natural killer cell platform for cancer immunotherapy
.
Mol Ther J Am Soc Gene Ther
2020
;
28
:
52
63
.
80.
Zhu
H
,
Blum
RH
,
Bernareggi
D
,
Ask
EH
,
Wu
Z
,
Hoel
HJ
, et al
Metabolic reprograming via deletion of CISH in human iPSC-derived NK cells promotes in vivo persistence and enhances anti-tumor activity
.
Cell Stem Cell
2020
;
27
:
224
37
.
81.
Daher
M
,
Basar
R
,
Gokdemir
E
,
Baran
N
,
Uprety
N
,
Nunez Cortes
AK
, et al
Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells
.
Blood
2020
;
doi: 10.1182/blood.2020007748. Epub ahead of print
.
82.
Ray
M
,
Lee
YW
,
Hardie
J
,
Mout
R
,
Yesilbag Tonga
G
,
Farkas
ME
, et al
CRISPRed macrophages for cell-based cancer immunotherapy
.
Bioconjug Chem
2018
;
29
:
445
50
.
83.
Kleinstiver
BP
,
Pattanayak
V
,
Prew
MS
,
Tsai
SQ
,
Nguyen
NT
,
Zheng
Z
, et al
High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects
.
Nature
2016
;
529
:
490
5
.
84.
Vakulskas
CA
,
Dever
DP
,
Rettig
GR
,
Turk
R
,
Jacobi
AM
,
Collingwood
MA
, et al
A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells
.
Nat Med
2018
;
24
:
1216
24
.
85.
Slaymaker
IM
,
Gao
L
,
Zetsche
B
,
Scott
DA
,
Yan
WX
,
Zhang
F
. 
Rationally engineered Cas9 nucleases with improved specificity
.
Science
2016
;
351
:
84
8
.
86.
Casini
A
,
Olivieri
M
,
Petris
G
,
Montagna
C
,
Reginato
G
,
Maule
G
, et al
A highly specific SpCas9 variant is identified by in vivo screening in yeast
.
Nat Biotechnol
2018
;
36
:
265
71
.
87.
Chen
JS
,
Dagdas
YS
,
Kleinstiver
BP
,
Welch
MM
,
Sousa
AA
,
Harrington
LB
, et al
Enhanced proofreading governs CRISPR-Cas9 targeting accuracy
.
Nature
2017
;
550
:
407
10
.
88.
Broeders
M
,
Herrero-Hernandez
P
,
Ernst
MPT
,
van der Ploeg
AT
,
Pijnappel
WWMP
. 
Sharpening the molecular scissors: advances in gene-editing technology
.
iScience
2020
;
23
:
100789
.
89.
Kantor
A
,
McClements
M
,
MacLaren
R
. 
CRISPR-Cas9 DNA base-editing and prime-editing
.
Int J Mol Sci
2020
;
21
:
6240
.
90.
Komor
AC
,
Kim
YB
,
Packer
MS
,
Zuris
JA
,
Liu
DR
. 
Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage
.
Nature
2016
;
533
:
420
4
.
91.
Nishida
K
,
Arazoe
T
,
Yachie
N
,
Banno
S
,
Kakimoto
M
,
Tabata
M
, et al
Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems
.
Science
2016
;
353
:
aaf8729
.
92.
Gaudelli
NM
,
Komor
AC
,
Rees
HA
,
Packer
MS
,
Badran
AH
,
Bryson
DI
, et al
Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage
.
Nature
2017
;
551
:
464
71
.
93.
Kuscu
C
,
Parlak
M
,
Tufan
T
,
Yang
J
,
Szlachta
K
,
Wei
X
, et al
CRISPR-STOP: gene silencing through base-editing-induced nonsense mutations
.
Nat Methods
2017
;
14
:
710
2
.
94.
Billon
P
,
Bryant
EE
,
Joseph
SA
,
Nambiar
TS
,
Hayward
SB
,
Rothstein
R
, et al
CRISPR-mediated base editing enables efficient disruption of eukaryotic genes through induction of STOP codons
.
Mol Cell
2017
;
67
:
1068
79
.
95.
Webber
BR
,
Lonetree
CL
,
Kluesner
MG
,
Johnson
MJ
,
Pomeroy
EJ
,
Diers
MD
, et al
Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors
.
Nat Commun
2019
;
10
:
5222
.
96.
Anzalone
AV
,
Randolph
PB
,
Davis
JR
,
Sousa
AA
,
Koblan
LW
,
Levy
JM
, et al
Search-and-replace genome editing without double-strand breaks or donor DNA
.
Nature
2019
;
576
:
149
57
.
97.
Schuster
A
,
Erasimus
H
,
Fritah
S
,
Nazarov
PV
,
van Dyck
E
,
Niclou
SP
, et al
RNAi/CRISPR screens: from a pool to a valid hit
.
Trends Biotechnol
2019
;
37
:
38
55
.
98.
Okada
M
,
Chikuma
S
,
Kondo
T
,
Hibino
S
,
Machiyama
H
,
Yokosuka
T
, et al
Blockage of core fucosylation reduces cell-surface expression of PD-1 and promotes anti-tumor immune responses of T cells
.
Cell Rep
2017
;
20
:
1017
28
.
99.
Shang
W
,
Jiang
Y
,
Boettcher
M
,
Ding
K
,
Mollenauer
M
,
Liu
Z
, et al
Genome-wide CRISPR screen identifies FAM49B as a key regulator of actin dynamics and T cell activation
.
Proc Natl Acad Sci U S A
2018
;
115
:
E4051
60
.
100.
Shang
W
,
Wang
F
,
Zhu
Q
,
Wang
L
,
Wang
H
. 
CRISPR/Cas9-based genetic screening to study T-cell function
.
Methods Mol Biol
2020
;
2111
:
59
70
.
101.
Ting
PY
,
Parker
AE
,
Lee
JS
,
Trussell
C
,
Sharif
O
,
Luna
F
, et al
Guide Swap enables genome-scale pooled CRISPR-Cas9 screening in human primary cells
.
Nat Methods
2018
;
15
:
941
6
.
102.
Shifrut
E
,
Carnevale
J
,
Tobin
V
,
Roth
TL
,
Woo
JM
,
Bui
CT
, et al
Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function
.
Cell
2018
;
175
:
1958
71
.
103.
Dong
MB
,
Wang
G
,
Chow
RD
,
Ye
L
,
Zhu
L
,
Dai
X
, et al
Systematic immunotherapy target discovery using genome-scale in vivo CRISPR screens in CD8 T cells
.
Cell
2019
;
178
:
1189
204
.
104.
Ye
L
,
Park
JJ
,
Dong
MB
,
Yang
Q
,
Chow
RD
,
Peng
L
, et al
In vivo CRISPR screening in CD8 T cells with AAV-Sleeping Beauty hybrid vectors identifies membrane targets for improving immunotherapy for glioblastoma
.
Nat Biotechnol
2019
;
37
:
1302
13
.
105.
Wei
J
,
Long
L
,
Zheng
W
,
Dhungana
Y
,
Lim
SA
,
Guy
C
, et al
Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy
.
Nature
2019
;
576
:
471
6
.
106.
Fraietta
JA
,
Nobles
CL
,
Sammons
MA
,
Lundh
S
,
Carty
SA
,
Reich
TJ
, et al
Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells
.
Nature
2018
;
558
:
307
12
.
107.
Nobles
CL
,
Sherrill-Mix
S
,
Everett
JK
,
Reddy
S
,
Fraietta
JA
,
Porter
DL
, et al
CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration
.
J Clin Invest
2020
;
130
:
673
85
.