Abstract
The advent of chimeric antigen receptor (CAR) engineering has led to the development of powerful cellular therapies for cancer. CAR-T cell based treatments have had notable clinical success, but logistical issues and associated toxicities are recognized limitations. There is an emerging interest in utilizing other immune effector cell types for CAR therapy. Natural killer cells are part of the innate immune system and these lymphocytes play a major role in immunosurveillance and anti-tumor immune responses. Incorporating chimeric antigen receptors into natural killer cells provides the opportunity to harness and enhance their innate cytotoxic potential towards malignancies. In this review we discuss the production of CAR engineered natural killer cells, highlight data on their preclinical and clinical efficacy, and examine the obstacles and strategies to overcome them.
Keywords: natural killer cells, chimeric antigen receptor, immunotherapy, resistance, clinical trial
Introduction
Cellular therapies for cancer have emerged as powerful treatment strategies that have helped revolutionize the immunotherapy field. The advent of chimeric antigen receptor (CAR) engineering, which was first applied in the 1990s for use in T cells with the goal of combining the beneficial anticancer characteristics of T cells with antigen specificity, has proven to be a successful example of personalized medicine.1 Clinical trials have shown particular promise in the use of CAR-T cells for the treatment of hematological B-cell malignancies2 and there are now five FDA-approved CAR-T cell therapies for hematologic cancers.3–9 Despite the impressive advances in the cellular therapy field over the past decade there are still major limitations and obstacles to the use of CAR-T cell based therapeutics. These drawbacks include: time-consuming production, the cumbersome logistics involving generation of autologous products for each patient, high cost, and a significant toxicity profile.10 Allogeneic CAR-T cells have been used to address some of these challenges, however this approach presents its own challenges, including graft versus host disease (GVHD) which poses a significant risk even when accounting for human leukocyte antigen (HLA) matching.11 Limiting side-effects include the substantial risk of cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS).12,13 The challenges to more widespread adoption of CAR-T cells in the clinic have spurred investigation of alternative immune effector cell populations as potential vehicles for CARs. In recent years, natural killer cells have emerged as promising candidates as an alternative to modified T cells.
Natural killer (NK) cells were first discovered in mice in 1975 and were shown to selectively kill leukemia cells.14 These lymphocytes are components of the innate immune system, and can eliminate cancerous cells without the need for prior sensitization, as their function and activity are regulated solely by the balance of signals received from activating and inhibitory receptors.15 NK cells mediate target cell lysis through several distinct mechanisms such as cytotoxic granule exocytosis, upregulation of death ligand expression (i.e., Fas and TRAIL), and production of cytokines such as interferon gamma (IFN-γ).16 NK cells are also capable of mediating antibody-dependent cell-mediated cytotoxicity (ADCC). In ADCC, monoclonal antibodies that target antigens expressed by tumor cells will recruit NK cells through FcγR-Fc interactions leading to NK cell activation and target cell lysis.17 NK cells have multiple advantages over T cells for CAR-based immunotherapy. As NK cell activation is triggered by their innate receptors, the addition of a CAR will redirect towards antigen-specific targets but NK cell-mediated cytotoxicity will be preserved even in the event of immune escape mechanisms such as target antigen loss or downregulation. Importantly, potential adverse events due to NK cell-based therapies are less likely due to their limited persistence in vivo and CRS, ICANS, and GVHD have not emerged as toxicities.18 Ultimately, CAR-NK cells are promising as the next generation of cellular therapy because they do not require HLA compatibility, are associated with minimal side-effects, and can be produced on a large scale from several sources to make “off-the-shelf” treatment a possibility. In this review we will briefly summarize CAR-NK cell design and discuss the progress being made in the development of NK cells as platforms for CAR-based therapies. We will focus on the challenges to CAR-NK cell therapy and the recent advances in improving their clinical efficacy.
CAR-NK Cell Production
Commonly used sources of functional NK cells for CAR-NK production include the NK-92 cell line, adult peripheral blood (AB), umbilical cord blood (CB), and induced pluripotent stem cells (iPSCs).19–21 Although autologous NK cells can be utilized to generate CAR-NK cells, patient sourced NK cells have limited efficacy against their cancer and enhancing their clinical utility with CAR engineering may be challenging.22 Depending on their origin, harvested NK cells have varying maturation stages, viability, and anti-tumor responses. The advantages and disadvantages associated with each source of NK cells are summarized in Figure 1. Some of the major benefits of utilizing NK-92 cells is they are highly cytotoxic, express relatively low levels of inhibitory receptors, and large numbers of these NK cells can quickly and easily be generated in vitro.23,24 However, as they were originally derived from a patient with a NK cell lymphoma, there is a small risk of tumor engraftment and irradiation prior to infusion is a safety requirement to prevent expansion. Unfortunately, this process negatively impacts their in vivo proliferation and persistence, and both factors have been shown to be crucial for the success of adoptive cellular therapy.25 An additional drawback is that NK-92 cells lack expression of the Fc-recognizing CD16 domain and are therefore unable to trigger ADCC without further manipulation. One of the downsides of using primary NK cells isolated from AB or CB is their in vitro culture and expansion is much more labor-intensive. However, both AB and CB have been shown to be an effective source of NK cells for CAR.26 Large numbers of NK cells can be obtained from samples of CB compared to AB; however, in contrast to AB, CAR-NK cells derived from CB progenitors mostly present an immature phenotype and have inferior cytotoxic capabilities prior to ex-vivo expansion.27 Generating clinical doses of CAR-NK cells using CB and AB NK cells is still a challenge and requires ex-vivo expansion of the cells. This ex-vivo expansion can be done through several methods.28 This includes coculturing with IL-2, IL-12, IL-15, IL-18 and/or IL-21.28 This method is relatively easy and has the benefit of not requiring feeder cells. However, it requires 4–10 weeks to reach clinically relevant numbers, which is significantly longer than utilizing feeder cells.29 CB and AB derived CAR-NK cells can also be expanded using universal antigen presenting cells (uAPC).30 AB and CB cells can be cocultured with the leukemia cell line K562, which is frequently transduced with membrane bound cytokines. Coculturing NK cells with HLA deficient K562 cells expressing CD48, 4–1BBL, and membrane-bound IL-21 causes the expansion of CAR-transduced CB and AB cells by >900-fold in only 2 weeks.30 NK cells derived from iPSCs offer some of the same benefits of NK-92, in that large numbers of mature and functional NK cells can be generated with relative ease.31 In contrast to NK-92 cells, iPSCs do not require irradiation and have also been shown to be effective upon CAR modification albeit with a less mature phenotype, reminiscent of CB-derived cells.32 Additionally, cytokine induced memory like (CIML) NK cells have been developed by activating AB cells with IL-12, IL-15, and IL-18.33 These cells exhibit potent antitumor responses and have been utilized in the treatment of leukemia, inducing complete remission in these patients.34 Recently, these cells have been transduced with CAR constructs targeted against CD19 and have effectively controlled lymphoma burden in vivo.35 These memory like CAR-NK cells exhibited increased degranulation and specific killing compared to conventional CAR-NK cells.35
A CAR construct is comprised of three components: an ectodomain responsible for antigen-recognition, a transmembrane linker, and an intracellular signaling domain. The extracellular antigen receptor portion is generally composed of a single-chain variable fragment (scFv). scFvs can target unique or overexpressed tumor cell antigens and CAR constructs are capable of targeting both surface proteins and HLA peptide complexes.36 When a CAR expressing NK cell binds the target antigen, the intracellular signaling domain transmits an activation signal, leading to cytokine secretion and lysis of the target cell.37 In first-generation CARs, the signaling domain was CD3ζ alone.38 Second-generation CARs added a single costimulatory domain such as CD28 or 4–1BB, while third-generation CARs contain two or more distinct costimulatory domains. Future generations of CARs are being developed with the goal of further modulating and enhancing the activation of CAR-engineered cells to kill malignant cells.39 A fourth generation of CARs was recently described, where the immune effector cells were endowed with two transgenic products, the CAR and a transgenic payload, such as the release of IL-7, IL-12, IL-15, IL-18, IL-23 or some combination.40,41 CAR constructs that implemented NK-specific 2B4, DAP-10, or DAP-12 co-stimulatory domains instead of CD28 or 4–1BB were characterized as having improved cytotoxicity and higher IFN-γ secretion.42 These CAR constructs are depicted in Figure 2. In order to introduce the CAR construct into a cell, genetically engineered viruses are used. CAR-NK cells are commonly generated through lentiviral or alpharetroviral transduction, with some reports of transduction efficiency well over 90% for both NK-92 and primary NK cells.43 Lentiviruses are capable of infecting both dividing and non-dividing cells and can be used for transduction of a wider variety of cell types including quiescent stem cells, providing an advantage over retroviral vectors.44 For a more detailed discussion of CAR-NK design and engineering see a review by Gong et al.45 CAR constructs enable NK cells to conjugate target cells more rapidly than unmodified primary NK cells or NK-92 cells, thereby enhancing their cytotoxic potential.46 In addition to the targeted cytotoxicity of CAR-NK cells, they may also still function through NK cell receptors in a CAR-independent manner. Some of the promising CAR targets under investigation with NK cells include CD38, CD20, mesothelin, CD22, CD19, HER2, and GD2.47 The best candidate antigens for use in CAR therapy will be minimally expressed in normal tissues, thus carrying a lower risk of off-target effects and maximizing clinical benefit while minimizing the risk of harmful toxicities to patients.
Pre-Clinical and Clinical Data on CAR-NK Cell Therapy
Pre-clinical data on the use of CAR-NK cells are very promising and have demonstrated efficacy for a variety of cancers and target antigens. CAR-NK cells derived from multiple sources are under extensive study for use in hematological malignancies and solid tumors.48 It is worth noting that although primary NK cells are an effective source for CAR modification, the majority of pre-clinical work has employed the NK-92 cell line. Second and third generation CAR-NK cells derived from NK-92 cells have shown to be effective in pre-clinical work targeting a range of antigens and cell types.49 CAR-NK-92 cells have enhanced cytotoxicity against T cell acute lymphoblastic leukemia and CD19+ leukemias and lymphomas, as well as HER2 overexpressing glioblastoma.50–52 Multiple studies have confirmed activity of CAR-NK-92 cells targeting ovarian cancer antigens in in vitro and in vivo pre-clinical models.53–55 Lastly, CAR-NK-92 also have pre-clinical efficacy when used to treat CS1 expressing multiple myeloma and prostate cancer.56,57 CAR-NK cells derived from the KHYG-1 have also been shown to have enhanced cytotoxic capabilities against glioblastoma cells expressing EGFRvIII.58 While there are relatively fewer studies employing CAR-NK cells sourced from AB, CB, or iPSCs; in vitro models and in vivo experiments have also demonstrated promising anti-tumor effects.59
While there have now been hundreds of clinical trials for CAR-T therapy, CAR-NK cell based treatments have only recently begun clinical investigation. There are currently only three published clinical trials on the use of CAR-NK therapy. In those clinical trials, the administration of CAR-NK cells were found to be safe and effective for patients with multiple myeloma, acute myeloid leukemia, and CD19+ lymphoid tumors.60–62 In a trial of eleven patients with either chronic lymphocytic leukemia (CLL) or non-Hodgkin’s lymphoma (NHL) who had previously received a median of four lines of therapy, a single infusion of anti-CD19 CAR-NK cells led to objective responses in eight patients, of whom 7 had a complete response within the first month after treatment.63 No serious adverse events were observed in any of the patients. Despite the limited number of published clinical trials on CAR-NK therapy, additional pilot clinical studies have achieved notable responses. Using an RNA electroporation approach, transient expression of a NKG2D RNA CAR significantly augmented the cytolytic activity of NK cells in vitro and was piloted as a treatment for two patients with chemotherapy-refractory metastatic colorectal cancer.64 Malignant cells in ascites fluid were almost undetectable after treatment and the observed volume of ascites was also significantly lower post-treatment with no recorded serious adverse effects. A third patient with metastatic colorectal cancer in the liver was treated by direct injection into liver lesions, which resulted in significant reduction in tumor burden. There are currently 32 CAR-NK based clinical trials registered as completed or active in NIH.gov (Table 1), with a majority ongoing in China. In summary, pre-clinical and early clinical data support further investigation of CAR-NK cell based therapeutics for a wide range of malignancies.
Table 1.
NCT Number | NK Cell Source | Target | Indication | Status |
---|---|---|---|---|
Hematological Malignancies | ||||
NCT00995137 | Haplo-identical donor | CD19 | ALL | Completed |
NCT02892695 | NK-92 | CD19 | CD19+ Leukemia/Lymphoma | Phase 1/2, Recruiting |
NCT02944162 | NK-92 | CD33 | AML | Phase 1/2, Recruiting |
NCT03940833 | NK-92 | BCMA | Multiple Myeloma | Phase 1/2, Recruiting |
NCT02742727 | NK-92 | CD7 | CD7+ Leukemia/Lymphoma | Phase 1/2, Recruiting |
NCT03056339 | Umbilical and Cord Blood | CD19 | B Cell Malignancies | Phase 1/2, Not yet recruiting |
NCT05092451 | Cord Blood | CD70 | B Cell Lymphoma, AML | Phase 1/2, Not yet recruiting |
NCT04887012 | Haplo-identical donor | CD19 | B Cell NHL | Phase 1, Recruiting |
NCT04796688 | AT19 | CD19 | B Cell Malignancies | Phase 1, Recruiting |
NCT05008536 | Umbilical and Cord Blood | BCMA | Multiple Myeloma | Phase 1, Recruiting |
NCT04796675 | Cord Blood | CD19 | B Cell Malignancies | Phase 1, Recruiting |
NCT05020678 | Allogeneic Donor | CD19 | B Cell Malignancies | Phase 1, Recruiting |
NCT04623944 | Allogeneic Donor | NKG2D | AML | Phase 1, Recruiting |
NCT05182073 | iPSC | BCMA | Multiple Myeloma | Phase 1, Recruiting |
NCT04245722 | iPSC | CD19 | B Cell Lymphoma, CLL | Phase 1, Recruiting |
NCT04555811 | iPSC | CD19 | B Cell Lymphoma | Phase 1, Recruiting |
NCT05247957 | Cord Blood | NKG2D | AML | Phase 1, Recruiting |
NCT05215015 | Undisclosed | CD33/CLL1 | AML | Phase 1, Recruiting |
NCT05008575 | Undisclosed | CD33 | AML | Phase 1, Recruiting |
NCT05379647 | Allogeneic Donor | CD19 | B Cell Malignancies | Phase 1. Recruiting |
NCT03824951 | iPSC | CD19 | B Cell Lymphoma | Phase 1, Not yet recruiting |
NCT03824964 | iPSC | CD19/CD22 | B Cell Lymphoma | Phase 1, Not yet recruiting |
NCT03692767 | iPSC | CD22 | B Cell Lymphoma | Phase 1, Not yet recruiting |
NCT03690310 | iPSC | CD19 | B Cell Lymphoma | Phase 1, Not yet recruiting |
NCT04639739 | Undisclosed | CD19 | NHL | Phase 1, Not yet recruiting |
Solid Tumors | ||||
NCT04390399 | t-haNK (NK-92 derived) | PD-L1 | Pancreatic Cancer | Phase 2, Recruiting |
NCT04847466 | t-haNK (NK-92 derived) | PD-L1 | Gastroesophageal Junction (GEJ) Cancers, Advanced HNSCC | Phase 2, Recruiting |
NCT02839954 | Undisclosed | MUC1 | MUC1+ Solid Tumors | Phase 1/2, Recruiting |
NCT03940820 | Undisclosed | ROBO1 | Solid Tumors | Phase 1/2, Recruiting |
NCT03931720 | Undisclosed | ROBO1 | Solid Tumors | Phase 1/2, Recruiting |
NCT03941457 | Undisclosed | ROBO1 | Pancreatic Cancer | Phase 1/2, Recruiting |
NCT03415100 | Allogeneic Donor | NKG2D | Metastatic Solid Tumors | Phase 1, Recruiting |
NCT03383978 | NK-92 | HER2 | Glioblastoma | Phase 1, Recruiting |
NCT05213195 | Undisclosed | NKG2D | Metastatic Colorectal Cancer | Phase 1, Recruiting |
NCT05194709 | Undisclosed | 5T4 | Solid tumors | Phase 1, Recruiting |
NCT03692663 | iPSC | PSMA | Prostate Cancer | Phase 1, Not yet recruiting |
NCT03692637 | iPSC | Mesothelin | Ovarian Cancer | Phase 1, Not yet recruiting |
NCT04050709 | t-haNK (NK-92 derived) | PD-L1 | Locally advanced or metastatic solid tumors | Phase 1, Not yet recruiting |
NHL, non-Hodgkin lymphoma; CLL, chronic lymphocytic leukemia; AML, acute myelogenous leukemia, HNSCC, head and neck squamous cell carcinoma
Challenges to CAR-NK Therapy and Strategies to Overcome Resistance
Pre-clinical and clinical data support the potential of CAR-NK cell therapies for cancer, however, a number of challenges to their clinical application must be addressed for that potential to be realized. First, the recent recognition of advantages that NK cells have over T cells as CAR engineering vehicles has led to the rapid development of CAR-NK therapeutics. Yet, the CAR constructs currently in use were designed for building CAR-T cells and recent evidence that the location of CAR binding epitopes modulates NK cell ability to bind antigen and become activated suggests such constructs may be suboptimal for application in NK cells.65 Designing CAR constructs specifically for NK cell activation and cytotoxicity could greatly improve efficacy. Another challenge is that, in contrast to T cells, NK cells are very sensitive to the freeze-thaw process. The survival rate and cytotoxic capabilities of CAR-NK cells are significantly reduced by thawing procedures, but functional activity may be restored by incubation with interleukin-2 (IL-2).66 Identification of ideal cryopreservation and recovery protocols will be necessary before CAR-NK cell therapy is viable as an “off-the-shelf” treatment.
One of the major barriers to CAR-NK cell therapy is the lack of persistence in vivo in the absence of cytokine support. While the lack of long-term proliferation in patients is desirable for safety, it is a limiting factor for efficacy. IL-2 and interleukin-15 (IL-15) are the two most commonly used cytokines to enhance the viability and prolong the persistence of adoptively NK cell therapies. The use of IL-2 is associated with substantial side effects and IL-15, which does not support Treg expansion and has a more favorable toxicity profile, is preferred.67 New CAR-NK cell engineering strategies incorporate genes in order to ectopically express cytokines such as IL-15 to improve the persistence of these cells in vivo.68 In order to address safety concerns, a suicide gene was also included which was able to quickly eliminate the CAR-modified NK cells. Furthermore, adoptive cellular therapies for solid tumors depend on proper homing and trafficking to the tumor site.69 Several studies have examined ways to engineer CAR-NK cells to make them more effective at infiltrating solid tumors. For example, NK cell homing to lymph nodes can be enhanced by the addition of the chemokine receptor CCR7 via trogocytosis.70 Similarly, electroporation with mRNA coding for CCR7 could improve NK cell migration towards lymph nodes that express the chemokine CCL19.71 Additionally, NK cells transduced with a CXCR2-encoding viral vector had augmented infiltration into renal cell carcinoma tumors that expressed CXCR2 cognate ligands.72 CXCR1, an interleukin-8 receptor, was found to be downregulated upon ex-vivo expansion of primary NK cells sourced from peripheral blood. After restoring expression through mRNA transfection, NK cells expressing an NKG2D CAR showed enhanced migration to and infiltration of solid tumors in mice with ovarian cancer xenografts.73 CAR-NK cells targeting EGFRvIII modified to overexpress the chemokine receptor CXCR4 showed greater tumor infiltration of glioblastoma secreting CXCL12/SDF-1α and likely enhanced migration to these tumors as well.74 Innovative strategies to overcome these challenges to CAR-NK cell therapy are being explored in numerous pre-clinical studies with planned evaluation of efficacy in upcoming clinical trials.
In addition to the above limitations of CAR-NK therapy, CAR-NK cells must also overcome tumor cell-based resistance mechanisms to NK cells. Resistance to NK cell therapy is complex and involves a multitude of factors that may be unique to individual patients.75 The addition of both first and second-generation CAR constructs targeting CD19 to NK cells has been shown to help overcome resistance to NK-killing in B-cell malignancies.76,77 These results demonstrate that the addition of CAR constructs alone may help overcome resistance. More novel CAR-NK cell based strategies aim to specifically tip the balance between NK cell activating and inhibitory receptor signals toward tumor-specific activation. For example, knocking out genes that code for proteins that transmit inhibitory signals may also enhance CAR-NK cell cytotoxicity and help overcome NK-cell resistance. Shp-2 knock-out in YT (NK-like) cells enhanced CAR-mediated cytotoxicity towards human prostate cancer cells resistant to NK-killing.78 Deletion of CISH, which codes for cytokine-inducible Src homology 2–containing protein and acts as an important cytokine checkpoint in NK cells, also resulted in greater expression of granzyme B, perforin, as well as other cytotoxicity and activation markers. CAR-NK cells with CISH KO constitutively expressing IL-15 doubled persistence time in vivo, enhanced metabolic fitness, and exhibited enhanced killing of CD19 positive lymphoma cells.79 Campana et al. designed a CAR construct that incorporated the two key signaling molecules, DAP10 and CD3ζ, and found markedly enhanced cytotoxic capabilities.80 Similarly, integration of the signaling lymphocyte activation molecule-related receptor 2B4 into CARs significantly enhanced NK cell activation and overcame NK cell resistance of target autologous leukemia cells.81 Additional resistance mechanisms that must be overcome involve the tumor microenvironment (TME). Hypoxia is one feature commonly observed in solid tumors that has a marked effect on NK cell function.82 Hypoxia has been shown to downregulate key NK activating receptors such as NKp46, NKp30, and NKG2D, resulting in decreased degranulation capability.83 Immunosuppressive cytokines such as TGF-β, which has been shown to suppress NK cell production of IFN-γ, reduce surface expression of NKp30 and NKG2D, and inhibit NK cell function are also found in the TME.84–86 Tumor-infiltrating NK cells isolated from patients with breast cancer have shown decreased expression of molecules associated with NK cytotoxicity including CD57, perforin, and granzyme B.87 CAR-NK cells can be engineered to render them resistant to the action of TGF-β. One promising strategy is the use of CRISPRCas9 technology to delete the TGF-β receptor 2 gene (TGFβR2) in primary human NK cells before CAR transduction, which renders them resistant to TGF-β mediated immunosuppression and enhanced their effector activity against a model of acute myeloid leukemia, as well as glioblastoma.88,89 In a similar strategy, CAR-NK cells were modified to express a dominant-negative TGF-β receptor, which binds TGF-β with high affinity but does not transduce downstream signaling, mitigating its suppressive effects and restored NK cell cytotoxicity.90 In order to effectively treat malignancies, especially solid tumors, CAR-NK cells must include rational designs to address the limitations of adoptive NK cell therapy as well as known resistance mechanisms to NK cell-based treatments.
Conclusion
Engineering strategies to introduce CARs to immune effector cells have resulted in potent additions to the immunotherapeutic arsenal. CAR-T cell therapy has led to remarkable outcomes in patients with relapsed or refractory hematological malignances and has begun to make inroads in the treatment of solid tumors. Unfortunately, there are a number of limitations to CAR-T therapy that are a barrier to clinical use for the majority of patients. CAR-NK cells are rapidly becoming a next-generation option for cellular immunotherapy. Combining the innate ability of natural killer cells to kill malignant cells with the targeted and enhanced cytotoxicity conveyed by CAR constructs has resulted in a technically feasible, effective, and safe therapy for a variety of cancers and antigen targets. While CAR-NK therapy still faces many challenges, pre-clinical studies of CAR-NK cells have provided a roadmap for the future development of engineering strategies to overcome these issues and increase clinical efficacy (Table 2). Tailoring CAR structures to NK cell biology will maximize their cytotoxic potential. In addition, optimization of protocols for the expansion and activation of NK cells for transfusion in patients will allow for the use of CAR-NK cells as an “off-the-shelf” treatment. Finally, additional alterations of NK cells beyond CAR transduction to improve trafficking and overcome the immunosuppressive TME will overcome current challenges. The growing number of CAR-NK cell clinical trials indicates that this promising therapy has only begun to have its transformative potential realized.
Table 2.
Challenge | Potential Solution |
---|---|
Current CAR constructs not optimal for NK cells | Design CAR constructs that specifically enhance NK cell activation and cytotoxicity. Incorporate activating receptors / signaling molecules such as DAP10, Dap12, CD3ζ, and 2B4 into CAR constructs65 |
High NK cell sensitivity to freeze-thaw process | Identification of optimal cryopreservation and recovery protocols66 |
Lack of persistence in vivo without cytokine support | CAR-NK modification for ectopic expression of cytokines such as IL-1567–69 |
Poor migration to and infiltration of solid tumors | CAR-NK modification to express chemokine receptors such as CCR7, CXCR1, CXCR2, CXCR470–74 |
Tumor cell-based resistance to NK cell-mediated lysis | Knock out genes that code for NK inhibitory receptors / proteins that play a role in inhibitory pathways such as CISH and Shp-278–79 |
The immunosuppressive nature of the TME | Mitigate the effects of immunosuppressive cytokines such as TGF-β through receptor deletion88–89 |
Highlights.
Natural Killer cells play a major role anti-tumor immune responses
Chimeric antigen receptor engineering has led to effective cellular immunotherapies
Natural Killer cells have several advantages over T cells for CAR-based therapy
Challenges to CAR-NK cells include trafficking and an immunosuppressive environment
CAR-NK cells are rapidly becoming a next-generation option for cellular therapy
Acknowledgements
This work was supported by NIH grants CA511008 (LMW) and CA50633 (LMW)
Declaration of Competing Interest
Dr. Louis Weiner is on the scientific advisory board for Jounce Therapeutics, Celldex Pharmaceuticals, Cytomx Therapeutics, Immunome Inc., Samyang Pharmaceuticals, Tessa Therapeutics, and Celularity Inc. Research collaborations funded by BioXcel Therapeutics.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.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 gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proceedings of the National Academy of Sciences. 1993;90(2):720–724. doi: 10.1073/pnas.90.2.720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hartmann J, Schüßler- Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells—challenges and opportunities in translating innovative treatment concepts. EMBO Molecular Medicine. 2017;9(9):1183–1197. doi: 10.15252/emmm.201607485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hansen DK, Dam M, Faramand RG. Toxicities associated with adoptive cellular therapies. Best Practice & Research Clinical Haematology. 2021;34(3):101287. doi: 10.1016/j.beha.2021.101287 [DOI] [PubMed] [Google Scholar]
- 4.Anagnostou T, Riaz IB, Hashmi SK, Murad MH, Kenderian SS. Anti-CD19 chimeric antigen receptor T-cell therapy in acute lymphocytic leukaemia: a systematic review and meta-analysis. The Lancet Haematology. 2020;7(11):e816–e826. doi: 10.1016/S2352-3026(20)30277-5 [DOI] [PubMed] [Google Scholar]
- 5.Porter DL, Hwang WT, Frey N v., et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Science Translational Medicine. 2015;7(303). doi: 10.1126/SCITRANSLMED.AAC5415/SUPPL_FILE/7-303RA139_TABLES_S6_TO_S11.ZIP [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bouchkouj N, Kasamon YL, de Claro RA, et al. FDA approval summary: Axicabtagene Ciloleucel for Relapsed or Refractory Large B-cell Lymphoma. Clinical Cancer Research. 2019;25(6):1702–1708. doi: 10.1158/1078-0432.CCR-18-2743/74253/AM/FDA-APPROVAL-SUMMARY-AXICABTAGENE-CILOLEUCEL-FOR [DOI] [PubMed] [Google Scholar]
- 7.Locke FL, Neelapu SS, Bartlett NL, et al. Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma. Molecular Therapy. 2017;25(1):285–295. doi: 10.1016/J.YMTHE.2016.10.020/ATTACHMENT/399FB790-E0B4-4D1A-AAA2-A6095F30F9B6/MMC1.PDF [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. New England Journal of Medicine. 2017;377(26):2531–2544. doi: 10.1056/NEJMOA1707447/SUPPL_FILE/NEJMOA1707447_DISCLOSURES.PDF [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Munshi NC, Anderson LD, Shah N, et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. New England Journal of Medicine. 2021;384(8):705–716. doi: 10.1056/NEJMOA2024850/SUPPL_FILE/NEJMOA2024850_DATA-SHARING.PDF [DOI] [PubMed] [Google Scholar]
- 10.Cerrano M, Ruella M, Perales MA, et al. The Advent of CAR T-Cell Therapy for Lymphoproliferative Neoplasms: Integrating Research Into Clinical Practice. Frontiers in Immunology. 2020;11. doi: 10.3389/fimmu.2020.00888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mehta RS, Rezvani K. Chimeric antigen receptor expressing natural killer cells for the immunotherapy of cancer. Frontiers in Immunology. 2018;9(FEB):1–12. doi: 10.3389/fimmu.2018.00283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Frey N, Porter D. Cytokine Release Syndrome with Chimeric Antigen Receptor T Cell Therapy. Biology of Blood and Marrow Transplantation. 2019;25(4):e123–e127. doi: 10.1016/j.bbmt.2018.12.756 [DOI] [PubMed] [Google Scholar]
- 13.Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Reviews. 2019;34:45–55. doi: 10.1016/j.blre.2018.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kiessling R. Natural Killer Cells in the Mouse. The Role of Products of the Histocompatibility Gene Complex in Immune Responses. Published online 1975:77–85. doi: 10.1016/b978-0-12-401660-6.50012-6 [DOI] [Google Scholar]
- 15.Rezvani K, Rouce R, Liu E, Shpall E. Engineering Natural Killer Cells for Cancer Immunotherapy. Molecular Therapy. 2017;25(8):1769–1781. doi: 10.1016/j.ymthe.2017.06.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sanchez CE, Dowlati EP, Geiger AE, et al. NK Cell Adoptive Immunotherapy of Cancer: Evaluating Recognition Strategies and Overcoming Limitations. Transplantation and Cellular Therapy. 2021;27(1):21–35. doi: 10.1016/j.bbmt.2020.09.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sondel PM, Alderson KL. Clinical cancer therapy by NK cells via antibody-dependent cell-mediated cytotoxicity. Journal of Biomedicine and Biotechnology. 2011;2011. doi: 10.1155/2011/379123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Liu E, Marin D, Banerjee P, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. New England Journal of Medicine. 2020;382(6):545–553. doi: 10.1056/NEJMoa1910607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nowakowska P, Romanski A, Miller N, et al. Clinical grade manufacturing of genetically modified, CAR-expressing NK-92 cells for the treatment of ErbB2-positive malignancies. Cancer Immunology, Immunotherapy. 2018;67(1):25–38. doi: 10.1007/s00262-017-2055-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Li Y, Hermanson DL, Moriarity BS, Kaufman DS. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell. 2018;23(2):181–192.e5. doi: 10.1016/j.stem.2018.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Herrera L, Juan M, Eguizabal C. Purification, Culture, and CD19- CAR Lentiviral Transduction of Adult and Umbilical Cord Blood NK Cells. Current Protocols in Immunology. 2020;131(1). doi: 10.1002/cpim.108 [DOI] [PubMed] [Google Scholar]
- 22.Liu E, Tong Y, Dotti G, et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia. 2018;32(2):520–531. doi: 10.1038/leu.2017.226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Maki G, Klingemann HG, Martinson JA, Tam YK. Factors Regulating the Cytotoxic Activity of the Human Natural Killer Cell Line, NK-92. Journal of Hematotherapy & Stem Cell Research. 2001;10(3):369–383. doi: 10.1089/152581601750288975 [DOI] [PubMed] [Google Scholar]
- 24.Tonn T, Schwabe D, Klingemann HG, et al. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy. 2013;15(12):1563–1570. doi: 10.1016/j.jcyt.2013.06.017 [DOI] [PubMed] [Google Scholar]
- 25.Besser MJ, Shapira-Frommer R, Treves AJ, et al. Clinical Responses in a Phase II Study Using Adoptive Transfer of Short-term Cultured Tumor Infiltration Lymphocytes in Metastatic Melanoma Patients. Clinical Cancer Research. 2010;16(9):2646–2655. doi: 10.1158/1078-0432.CCR-10-0041 [DOI] [PubMed] [Google Scholar]
- 26.Herrera L, Santos S, Vesga MA, et al. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Scientific Reports. 2019;9(1). doi: 10.1038/s41598-019-55239-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Luevano M, Daryouzeh M, Alnabhan R, et al. The unique profile of cord blood natural killer cells balances incomplete maturation and effective killing function upon activation. Human Immunology. 2012;73(3):248–257. doi: 10.1016/j.humimm.2011.12.015 [DOI] [PubMed] [Google Scholar]
- 28.Gong Y, Klein Wolterink RGJ, Wang J, Bos GMJ, Germeraad WTV. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. Journal of Hematology & Oncology 2021 14:1. 2021;14(1):1–35. doi: 10.1186/S13045-021-01083-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lapteva N, Szmania SM, van Rhee F, Rooney CM. Clinical grade purification and expansion of natural killer cells. Crit Rev Oncog. 2014;19(1–2):121–132. doi: 10.1615/CRITREVONCOG.2014010931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Konrad Odendahl M, Red Cross Blood Donor Services G, Leonid Metelitsa G, et al. GMP-Compliant Universal Antigen Presenting Cells (uAPC) Promote the Metabolic Fitness and Antitumor Activity of Armored Cord Blood CAR-NK Cells. Frontiers in Immunology | www.frontiersin.org. 2021;12:626098. doi: 10.3389/fimmu.2021.626098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Knorr DA, Ni Z, Hermanson D, et al. Clinical-Scale Derivation of Natural Killer Cells From Human Pluripotent Stem Cells for Cancer Therapy. Stem Cells Translational Medicine. 2013;2(4):274–283. doi: 10.5966/sctm.2012-0084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hermanson DL, Bendzick L, Pribyl L, et al. Induced Pluripotent Stem Cell-Derived Natural Killer Cells for Treatment of Ovarian Cancer. Stem Cells. 2016;34(1):93–101. doi: 10.1002/stem.2230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cooper MA, Elliott JM, Keyel PA, Yang L, Carrero JA, Yokoyama WM. Cytokine-induced memory-like natural killer cells. Proc Natl Acad Sci U S A. 2009;106(6):1915. doi: 10.1073/PNAS.0813192106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Romee R, Rosario M, Berrien-Elliott MM, et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci Transl Med. 2016;8(357):357ra123. doi: 10.1126/SCITRANSLMED.AAF2341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gang M, Marin ND, Wong P, et al. CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas. Blood. 2020;136(20):2308. doi: 10.1182/BLOOD.2020006619 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hermanson DL, Kaufman DS. Utilizing chimeric antigen receptors to direct natural killer cell activity. Frontiers in Immunology. 2015;6(APR). doi: 10.3389/fimmu.2015.00195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang W, Jiang J, Wu C. CAR-NK for tumor immunotherapy: Clinical transformation and future prospects. Cancer Letters. 2020;472(September 2019):175–180. doi: 10.1016/j.canlet.2019.11.033 [DOI] [PubMed] [Google Scholar]
- 38.Finney HM, Akbar AN, Lawson ADG. Activation of Resting Human Primary T Cells with Chimeric Receptors: Costimulation from CD28, Inducible Costimulator, CD134, and CD137 in Series with Signals from the TCRζ Chain. The Journal of Immunology. 2004;172(1):104–113. doi: 10.4049/jimmunol.172.1.104 [DOI] [PubMed] [Google Scholar]
- 39.Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discovery. 2013;3(4):388–398. doi: 10.1158/2159-8290.CD-12-0548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opinion on Biological Therapy. 2015;15(8):1145–1154. doi: 10.1517/14712598.2015.1046430 [DOI] [PubMed] [Google Scholar]
- 41.Chmielewski M, Abken H. TRUCKS, the fourth-generation CAR T cells: Current developments and clinical translation. Advances in Cell and Gene Therapy. 2020;3(3):e84. doi: 10.1002/ACG2.84 [DOI] [Google Scholar]
- 42.Wrona E, Borowiec M, Potemski P. CAR-NK Cells in the Treatment of Solid Tumors. International Journal of Molecular Sciences. 2021;22(11):5899. doi: 10.3390/ijms22115899 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Suerth JD, Morgan MA, Kloess S, et al. Efficient generation of gene-modified human natural killer cells via alpharetroviral vectors. Journal of Molecular Medicine. 2016;94(1):83–93. doi: 10.1007/s00109-015-1327-6 [DOI] [PubMed] [Google Scholar]
- 44.van Til NP, Wagemaker G. Lentiviral Gene Transduction of Mouse and Human Hematopoietic Stem Cells. In:; 2014:311–319. doi: 10.1007/978-1-4939-1133-2_21 [DOI] [PubMed] [Google Scholar]
- 45.Gong Y, Klein Wolterink RGJ, Wang J, Bos GMJ, Germeraad WT V. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. Journal of Hematology & Oncology. 2021;14(1):73. doi: 10.1186/s13045-021-01083-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ravi D, Sarkar S, Purvey S, et al. Interaction kinetics with transcriptomic and secretory responses of CD19-CAR natural killer-cell therapy in CD20 resistant non-hodgkin lymphoma. Leukemia. 2020;34(5):1291–1304. doi: 10.1038/s41375-019-0663-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.MacKay M, Afshinnekoo E, Rub J, et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nature Biotechnology. 2020;38(2):233–244. doi: 10.1038/s41587-019-0329-2 [DOI] [PubMed] [Google Scholar]
- 48.Kailayangiri S, Altvater B, Spurny C, et al. Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA-G. OncoImmunology. 2017;6(1):e1250050. doi: 10.1080/2162402X.2016.1250050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Pinz KG, Yakaboski E, Jares A, et al. Targeting T-cell malignancies using anti-CD4 CAR NK-92 cells. Oncotarget. 2017;8(68):112783–112796. doi: 10.18632/oncotarget.22626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.You F, Wang Y, Jiang L, et al. A novel CD7 chimeric antigen receptor-modified NK-92MI cell line targeting T-cell acute lymphoblastic leukemia. Am J Cancer Res. 2019;9(1):64–78. [PMC free article] [PubMed] [Google Scholar]
- 51.Oelsner S, Friede ME, Zhang C, et al. Continuously expanding CAR NK-92 cells display selective cytotoxicity against B-cell leukemia and lymphoma. Cytotherapy. 2017;19(2):235–249. doi: 10.1016/j.jcyt.2016.10.009 [DOI] [PubMed] [Google Scholar]
- 52.Zhang C, Burger MC, Jennewein L, et al. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. JNCI: Journal of the National Cancer Institute. 2016;108(5). doi: 10.1093/jnci/djv375 [DOI] [PubMed] [Google Scholar]
- 53.Klapdor R, Wang S, Morgan M, et al. Characterization of a Novel Third-Generation Anti-CD24-CAR against Ovarian Cancer. International Journal of Molecular Sciences. 2019;20(3):660. doi: 10.3390/ijms20030660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ao X, Yang Y, Li W, et al. Anti-αFR CAR-engineered NK-92 Cells Display Potent Cytotoxicity Against αFR-positive Ovarian Cancer. Journal of Immunotherapy. 2019;42(8):284–296. doi: 10.1097/CJI.0000000000000286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cao B, Liu M, Wang L, et al. Use of chimeric antigen receptor NK-92 cells to target mesothelin in ovarian cancer. Biochemical and Biophysical Research Communications. 2020;524(1):96–102. doi: 10.1016/j.bbrc.2020.01.053 [DOI] [PubMed] [Google Scholar]
- 56.Chu J, Deng Y, Benson DM, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2014;28(4):917–927. doi: 10.1038/leu.2013.279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Montagner IM, Penna A, Fracasso G, et al. Anti-PSMA CAR-Engineered NK-92 Cells: An Off-the-Shelf Cell Therapy for Prostate Cancer. Cells. 2020;9(6):1382. doi: 10.3390/cells9061382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Murakami T, Nakazawa T, Natsume A, et al. Novel human NK cell line carrying CAR targeting EGFRvIII induces antitumor effects in glioblastoma cells. Anticancer Research. 2018;38(9):5049–5056. doi: 10.21873/anticanres.12824 [DOI] [PubMed] [Google Scholar]
- 59.Wang W, Jiang J, Wu C. CAR-NK for tumor immunotherapy: Clinical transformation and future prospects. Cancer Letters. 2020;472:175–180. doi: 10.1016/j.canlet.2019.11.033 [DOI] [PubMed] [Google Scholar]
- 60.Jiang H, Zhang W, Shang P, et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Molecular Oncology. 2014;8(2):297–310. doi: 10.1016/j.molonc.2013.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tang X, Yang L, Li Z, et al. First-in-man clinical trial of CAR NK-92 cells: safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am J Cancer Res. 2018;8(6):1083–1089. [PMC free article] [PubMed] [Google Scholar]
- 62.Liu E, Marin D, Banerjee P, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. New England Journal of Medicine. 2020;382(6):545–553. doi: 10.1056/NEJMoa1910607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liu E, Marin D, Banerjee P, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. New England Journal of Medicine. 2020;382(6):545–553. doi: 10.1056/NEJMOA1910607/SUPPL_FILE/NEJMOA1910607_DATA-SHARING.PDF [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Xiao L, Cen D, Gan H, et al. Adoptive Transfer of NKG2D CAR mRNA-Engineered Natural Killer Cells in Colorectal Cancer Patients. Molecular Therapy. 2019;27(6):1114–1125. doi: 10.1016/j.ymthe.2019.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Siegler EL, Zhu Y, Wang P, Yang L. Off-the-Shelf CAR-NK Cells for Cancer Immunotherapy. Cell Stem Cell. 2018;23(2):160–161. doi: 10.1016/j.stem.2018.07.007 [DOI] [PubMed] [Google Scholar]
- 66.Voshol H, Dullens HFJ, Otter W Den, Vliegenthart JFG. Human natural killer cells: a convenient purification procedure and the influence of cryopreservation on cytotoxic activity. Journal of Immunological Methods. 1993;165(1):21–30. doi: 10.1016/0022-1759(93)90102-D [DOI] [PubMed] [Google Scholar]
- 67.Conlon KC, Lugli E, Welles HC, et al. Redistribution, Hyperproliferation, Activation of Natural Killer Cells and CD8 T Cells, and Cytokine Production During First-in-Human Clinical Trial of Recombinant Human Interleukin-15 in Patients With Cancer. Journal of Clinical Oncology. 2015;33(1):74–82. doi: 10.1200/JCO.2014.57.3329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang X, Jasinski DL, Medina JL, Spencer DM, Foster AE, Bayle JH. Inducible MyD88/CD40 synergizes with IL-15 to enhance antitumor efficacy of CAR-NK cells. Blood Advances. 2020;4(9):1950–1964. doi: 10.1182/bloodadvances.2020001510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Halama N, Braun M, Kahlert C, et al. Natural Killer Cells are Scarce in Colorectal Carcinoma Tissue Despite High Levels of Chemokines and Cytokines. Clinical Cancer Research. 2011;17(4):678–689. doi: 10.1158/1078-0432.CCR-10-2173 [DOI] [PubMed] [Google Scholar]
- 70.Somanchi SS, Somanchi A, Cooper LJN, Lee DA. Engineering lymph node homing of ex vivo–expanded human natural killer cells via trogocytosis of the chemokine receptor CCR7. Blood. 2012;119(22):5164–5172. doi: 10.1182/blood-2011-11-389924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Carlsten M, Levy E, Karambelkar A, et al. Efficient mRNA-Based Genetic Engineering of Human NK Cells with High-Affinity CD16 and CCR7 Augments Rituximab-Induced ADCC against Lymphoma and Targets NK Cell Migration toward the Lymph Node-Associated Chemokine CCL19. Frontiers in Immunology. 2016;7. doi: 10.3389/fimmu.2016.00105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kremer V, Ligtenberg MA, Zendehdel R, et al. Genetic engineering of human NK cells to express CXCR2 improves migration to renal cell carcinoma. Journal for ImmunoTherapy of Cancer. 2017;5(1):73. doi: 10.1186/s40425-017-0275-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ng YY, Tay JCK, Wang S. CXCR1 Expression to Improve Anti-Cancer Efficacy of Intravenously Injected CAR-NK Cells in Mice with Peritoneal Xenografts. Molecular Therapy - Oncolytics. 2020;16:75–85. doi: 10.1016/j.omto.2019.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Müller N, Michen S, Tietze S, et al. Engineering NK cells modified with an EGFRvIII-specific chimeric antigen receptor to overexpress CXCR4 improves immunotherapy of CXCL12/SDF-1α-secreting glioblastoma. Journal of Immunotherapy. 2015;38(5):197–210. doi: 10.1097/CJI.0000000000000082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sordo-Bahamonde C, Vitale M, Lorenzo-Herrero S, López-Soto A, Gonzalez S. Mechanisms of resistance to NK cell immunotherapy. Cancers (Basel). 2020;12(4). doi: 10.3390/cancers12040893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Romanski A, Uherek C, Bug G, et al. CD 19- CAR engineered NK - 92 cells are sufficient to overcome NK cell resistance in B- cell malignancies. Journal of Cellular and Molecular Medicine. 2016;20(7):1287–1294. doi: 10.1111/jcmm.12810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106(1):376–383. doi: 10.1182/blood-2004-12-4797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Subrakova VG, Kulemzin SV., Belovezhets TN, et al. shp-2 gene knockout upregulates CAR-driven cytotoxicity of YT NK cells. Vavilov Journal of Genetics and Breeding. 2020;24(1):80–86. doi: 10.18699/VJ20.598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Daher M, Basar R, Gokdemir E, et al. Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells. Blood. 2021;137(5):624–636. doi: 10.1182/blood.2020007748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chang YH, Connolly J, Shimasaki N, Mimura K, Kono K, Campana D. A Chimeric Receptor with NKG2D Specificity Enhances Natural Killer Cell Activation and Killing of Tumor Cells. Cancer Research. 2013;73(6):1777–1786. doi: 10.1158/0008-5472.CAN-12-3558 [DOI] [PubMed] [Google Scholar]
- 81.Altvater B, Landmeier S, Pscherer S, et al. 2B4 (CD244) Signaling by Recombinant Antigen-specific Chimeric Receptors Costimulates Natural Killer Cell Activation to Leukemia and Neuroblastoma Cells. Clinical Cancer Research. 2009;15(15):4857–4866. doi: 10.1158/1078-0432.CCR-08-2810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Muz B, de la Puente P, Azab F, Azab AK. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia. Published online December 2015:83. doi: 10.2147/HP.S93413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Balsamo M, Manzini C, Pietra G, et al. Hypoxia downregulates the expression of activating receptors involved in NK-cell-mediated target cell killing without affecting ADCC. European Journal of Immunology. 2013;43(10):2756–2764. doi: 10.1002/eji.201343448 [DOI] [PubMed] [Google Scholar]
- 84.Laouar Y, Sutterwala FS, Gorelik L, Flavell RA. Transforming growth factor-β controls T helper type 1 cell development through regulation of natural killer cell interferon-γ. Nature Immunology. 2005;6(6):600–607. doi: 10.1038/ni1197 [DOI] [PubMed] [Google Scholar]
- 85.Castriconi R, Cantoni C, Della Chiesa M, et al. Transforming growth factor β1 inhibits expression of NKP30 and NKG2d receptors: Consequences for the NK-mediated killing of dendritic cells. Proc Natl Acad Sci U S A. 2003;100(7):4120–4125. doi: 10.1073/pnas.0730640100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Batlle E, Massagué J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity. 2019;50(4):924–940. doi: 10.1016/j.immuni.2019.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mamessier E, Sylvain A, Thibult ML, et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. Journal of Clinical Investigation. 2011;121(9):3609–3622. doi: 10.1172/JCI45816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Daher M, Basar R, Shaim H, et al. The TGF-β/SMAD Signaling Pathway As a Mediator of NK Cell Dysfunction and Immune Evasion in Myelodysplastic Syndrome. Blood. 2017;130(Supplement 1):53. [Google Scholar]
- 89.Shaim H, Shanley M, Basar R, et al. Targeting the αv integrin/TGF-β axis improves natural killer cell function against glioblastoma stem cells. The Journal of Clinical Investigation. 2021;131(14). doi: 10.1172/JCI142116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yvon ES, Burga R, Powell A, et al. Cord blood natural killer cells expressing a dominant negative TGF-β receptor: Implications for adoptive immunotherapy for glioblastoma. Cytotherapy. 2017;19(3):408–418. doi: 10.1016/j.jcyt.2016.12.005 [DOI] [PubMed] [Google Scholar]