Open access peer-reviewed chapter

Cancer Immunotherapy and Cytotoxicity: Current Advances and Challenges

Written By

Leisheng Zhang and Hui Cai

Submitted: 25 March 2022 Reviewed: 05 May 2022 Published: 12 June 2022

DOI: 10.5772/intechopen.105184

From the Edited Volume

Cytotoxicity - Understanding Cellular Damage and Response

Edited by Anil Sukumaran and Mahmoud Ahmed Mansour

Chapter metrics overview

103 Chapter Downloads

View Full Metrics

Abstract

Immunotherapies are revolutionizing strategies for cancer treatment and infectious disease administration, which thus occupy worldwide concerns and enthusiasms for conquering relapsing and refractory immunodysfuction-related diseases. Current preclinical and clinical studies have suggested the partial success and promising potential of cancer management by various immunotherapies such as cancer vaccine, lymphocyte-promoting cytokines, checkpoint inhibitors and the cellular immunotherapy. However, the precise controlled modulation of the recipient’s immune system as well as the concomitant cytotoxicity remains the core challenge in the broad implementation of cancer immunotherapies. In this Chapter, we mainly focus on the latest updates of the cytotoxicity of cancer immunocytotherapy, together with the remarkable opportunities and conspicuous challenges, which represent the paradigm for boosting the immune system to enhance antitumor responses and ultimately eliminate malignancies. Collectively, we summarize and highlight the auspicious improvement in the efficacy and cytotoxicity of cancer immunotherapy and will benefit the large-scale preclinical investigations and clinical practice in adoptive immunotherapy.

Keywords

  • cancer immunotherapy
  • immune cells
  • hematopoietic malignancies
  • metastatic solid tumors
  • cytotoxicity

1. Introduction

To date, thousands upon thousands of people suffer and even die from various tumors with high morbidity and mortality including malignant hematologic tumors and metastatic solid tumors worldwide, which have cause tremendous physical and mental stress to the patients and their guardians [1, 2, 3, 4, 5, 6]. Despite the encouraging progresses in cancer treatment, one of the core dilemmas is the current limitation in classical therapeutic modalities such as surgery, radiation and chemotherapy [7, 8]. For instance, surgery in combination with radiation treatment and chemotherapy drugs has been proved to be effective for localized cancers without metastasis and diffusion [9, 10]. Chemoradiotherapy has been considered as a synergistic anticancer remedy for locally advanced solid tumors whereas with increased damage to normal tissues and microbiota resistance [11, 12, 13]. Distinguish from the conventional cancer treatment (e.g., surgery, radiotherapy and chemotherapy), noncellular immunotherapy such as checkpoint inhibitors (e.g., PD-1, PD-L1, CTLA4), lymphocyte-promoting cytokines (e.g., IFN-γ, GM-CSF, G-CSF), and cancer vaccines (e.g., mRNAs) has been continuously developed to fulfill the goals for cancer administration as well [14, 15, 16, 17]. Additionally, current progress has also highlighted the feasibility of nanomaterials (e.g., organic nanomaterials, inorganic nanomaterials, organic–inorganic hybrid nanomaterials) as promising agents for cancer therapy based on the knowledge of nanobiotechnology and clinical biomedicine [18, 19, 20]. However, the significant disadvantages of the aforementioned strategies are apparent and should not be ignored including drug delivery barriers, graft-versus-host disease, off-target effects and severe toxicity [5, 21, 22].

Therewith, the assumption of eradicating tumors by utilizing the human immune system has been successfully and extensively practiced for the past decades by pioneering investigators and clinicians [23]. To date, a series of cellular Immunotherapy has been identified to promote the outcomes and reduce adverse reactions of cancer patients, and in particular, those with late-stage patients with various treatment-refractory cancers [14, 15, 16, 17]. Therefore, in this chapter, we mainly focus on the progress as well as the prospective and challenges in cellular immunotherapy for cancer treatment including the patient-specific immune cells (e.g., tumor infiltrating lymphocytes, cytokine induced killer cells), innate immunocytes (e.g., natural killer cells, dendritic cells, macrophage), adaptive immunocytes (T lymphocytes, B lymphocytes), engineered immunocytes (e.g., chimeric antigen receptor-transduced T cells, T cell receptor-transduced T cells, CAR-NK, CAR-M) [21, 24, 25, 26, 27]. Collectively, the immune cell-mediated cancer immunotherapy has constituted a promising area of cancer biotherapy.

Advertisement

2. Classification of immunocytes

2.1 Patient-associated immunocytes

2.1.1 Lymphokine-activated killer cells (LAKs)

LAKs are effector cells with significant cytotoxic activity, which are induced by culturing with recombinant IL-2 and have been proven effective against NK cell-resistant allogeneic and autologous tumor cells or cell lines [28]. Generally, LAKs after regional intra-arterial perfusion mainly accumulate at tumor sites, whereas the intravenously infused LAKs first accumulate in lung tissues before migrating to the liver regions [29].

LAKs-mediated adoptive immunotherapy has been confirmed with limited efficacy in vivo ranging from 20–30% including the metastatic or advanced tumor models and clinical studies upon patients (e.g., hepatic carcinoma, renal cell carcinoma, melanoma, leukemia) [30, 31, 32]. For example, Rosenberg et al. reported the treatment of 157 patients with advanced cancer using LAKs and IL-2, and confirmed the marked tumor regression and even remission by the immunotherapeutic approach. Nevertheless, the ultimate role of LAKs-based cancer immunotherapy still awaits further improvement including the efficacy and the decrease of toxicity and complexity [32].

2.1.2 Tumor-infiltrating lymphocytes (TILs)

TILs are infiltrating lymphocytes in cancer tissues and play a critical role in mediating response to chemotherapy [33]. To date, of the tumor-infiltrating immune cells such as mast cells, macrophages, dendritic cells and leucocytes, TILs have been considered as selected heterogeneous populations of T lymphocytes with a higher and specific immunological reactivity against cancer cells than the non-infiltrating subset [34]. Despite the accumulation of TILs has been recognized as prognosis for elevated survival and clinical outcomes, yet tumors with high level of TILs also with increased PD-1 immune checkpoint expression [35]. Thus, the feasibility of TILs as biomarker for reflecting the immune responses and predicting the clinical outcomes of cancer immunotherapy still need systematic formulations and detailed explorations [34]. Generally, although ineffective for in vivo tumor elimination, yet TILs are adequate to functionate proliferation and effector capacity when separated from immunosuppressive tumor microenvironment [36]. Taken together, identification of specific subpopulations and the corresponding molecular mechanism of TILs in cancer might be of great importance for guiding prognosis and developing appropriate sequencing of immunotherapy [33].

2.1.3 Cytokine-induced killer cells (CIKs)

CIKs are a heterogeneous population of CD3+CD56+ natural killer T (NKT) cells and recognized as pharmacological tools for tumor immunotherapy, and in particular, in refractory to conventional radiotherapy and chemotherapy [37, 38]. Generally, CIKs contain two main subsets including the CD3 + CD56+ and CD3 + CD56- subpopulations endowed with higher anti-tumor activity and higher proliferation ability, respectively. Mature CIKs express active receptors of NK cells including NKG2D and DNAM-1, whereas with minimal expression of NKG2A, NKp44, NKp46 or CD94 [39].

For decades, numerous clinical trials of CIKs-based tumor immunotherapy have been registered attribute to the distinctive properties including intense MHC-independent antitumor activity, low toxicity effects and high safety on healthy cells [39, 40]. For instance, Yu et al. verified that CIKs-based immunotherapy was an effective adjuvant strategy in the early stage of hepatocellular carcinoma (HCC) rather than advanced HCC, and suggested that targeting myeloid-derived suppressor cells (MDSCs) would provide additional therapeutic benefits alongside CIKs-based therapy [23]. Moreover, CIKs can be expanded to match the clinical relevant rates cost-effectively and conveniently by a simple protocol, which is also the key issue for clinical application of adoptive immunotherapy [39].

Of the multiple modalities for cancer immunotherapy, vaccination with DC-CIKs has revealed limited therapeutic success in the administration of advanced solid tumors [41]. Therefore, considering the shortcomings of CIKs-based cellular immunotherapy alone, integrated therapy or combined modality therapy such as CIKs, cytokines, gene editing, immune checkpoints, radiotherapy and chemotherapy have better potentiality in relieving the major side effects and improving the clinical outcomes of standard treatment options [40].

2.2 Innate immunocytes

2.2.1 Natural killer (NK) cells

NK cells are heterogeneous cell population with unique characteristics of and belong to the innate lymphoid cells (ILCs), which can be divided into the cytotoxic CD56dimCD16high and IFN-γ-producing CD56brightCD16low/neg subsets and play an important role in both innate and adoptive immune responses dispense with preliminary antigen presentation via receptor-ligand mediated cytotoxicity, antibody-dependent cell-mediated cytotoxicity (ADCC), release of perforin and granzyme as well as cytokine-based paracrine effects (e.g., IFN-γ, GM-CSF) [21, 42].

For decades, we and other investigators have reported the generation of NK cells from various sources such as cell lines (e.g., NK-92, NK-92MI, YT), perinatal blood (e.g., cord blood, placental blood), peripheral blood, hematopoietic stem cells (HSCs) and human pluripotent stem cells (e.g., human embryonic stem cells, human induced pluripotent stem cells) [21, 43, 44, 45, 46, 47, 48]. Distinguish from the chimeric antigen receptor-transduced T cells (CAR-T), NK cells with non-MHC-restricted recognition revealed reliable cytotoxicity against pathogenic microorganism and tumor cells without the significant disadvantages graft-versus-host disease (GvHD), cytokine release syndrome (CRS) and neurotoxicity [49, 50, 51]. Moreover, NK cells are supposed to eliminate non-proliferating or quiescent cancer stem cells (CSCs), which collectively highlights the possibility and preponderance of NK cell-based cytotherapy for cancer immunosurveillance and immunotherapy [52, 53].

2.2.2 Dendritic cells (DCs)

DCs are most potent antigen-presenting cells (APCs) arising from lympho-myeloid hematopoiesis and linking the innate and adaptive immunity, which are firstly identified in 1973 and capable of initiating and activating lymphocytes including T cells and B cells and thus play a unique function in cancer immunotherapy as well as the tumor microenvironment [54, 55, 56, 57]. As recently reviewed by Stevens and the colleagues, the advent of DC-based immunotherapy and immune checkpoint inhibitors (e.g., PD-1/PD-L1, CTLA-4) has become a paradigm shift in the treatment of cancers including the small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) [58].

Notably, DCs derived in vitro or naturally circulating DCs loaded with the tumor-associated antigens (TAAs) are safe and feasible for eliciting a robust and tumor-directed immune response [59]. Briefly, immature DCs are attracted by the damage-associated molecular patterns (DAMPs) and then transit to a mature phenotype with capabilities of phagocytic cargo processing and antigenic component engulfment [60]. Additionally, DCs have been reported with synergistic effect with CIKs or thoracic radiotherapy for the management of locally advanced or metastatic NSCLCs [61, 62].

2.2.3 Macrophages (MΦ)

Macrophages (MΦ), one type of innate immune cells including the M1 (inhibit growth and kill) and M2 (promote growth and repair) subtypes, possess superior antineoplastic effects due to the phenomena of engulfing pathogens and dead cells, which are pivotal modulators of tissue regeneration and hematopoietic stem cell renewal as well as cancer progression [63, 64, 65, 66]. However, considering the purposiveness of identification of tumor-specific anticancer responses, the characteristics of MΦ in cancer as bidirectional executors have been largely overlooked for a long period [67, 68].

Current updates have indicated the biofunction of macrophages in the regression of cancers by modulating the polarization peculiarity of the predominate M2 repair-type MΦ (pro-tumor) to M1 kill-type (anti-tumor) [69]. Disturbances in MΦ function usually result in deficiency of anti-inflammatory MΦ, uncontrolled secretion of inflammatory factors (e.g., TGF-β, VEGF, EGF, PGE2, IFN-γ) and production of nitric oxide (NO) as well as poor communication with functional cells (e.g., endothelial cells, epithelial cells, lymphocytes) and cancer cells [70, 71]. For instance, the tumor-associated MΦ (TAMs) are recognized as the prominent components in tumor microenvironment (TME), which thus considered as promising and advantaged targets for developing immunotherapeutic strategy [72, 73].

Thus, MΦ/innate immunology can be orchestrated to play a critical role in indirectly or directly combating numerous hematological malignancies and metastatic solid tumors via the activity of M1 kill-type and the stimulatory effect of M1-type MΦ to cytotoxic Th1 cells and relative effector cells, respectively [69, 74]. Moreover, state-of-the-art updates have suggested the feasibility of conversion of the dominating cancer growth-promoting MΦ into growth-inhibiting type and the resultant progression in cancer immunotherapy as well [69].

2.3 Adaptive immunocytes

2.3.1 T lymphocytes

According to the tumor immunosurveillance theory, considerable attention has been caused on enhancing the effectiveness and cytotoxicity of antitumor immunity, and in particular, the T lymphocyte-based host immune response and the accompanied T cell signaling and metabolism as well [75]. T lymphocytes are CD3+ mononuclear cells and are essential for eradicating tumor cells, allergens and microorganisms as well as tissue repair, and thus people with the impairment or dysfunction of T cells are at high risk of cancers and infections and eventually poor prognosis or mortality [76, 77].

Immunotherapeutic resistance remains the substantial barrier of cancer immunotherapy, and the selection of appropriate standard-of-care treatments is critical for combating tumors in preclinical and clinical studies [78, 79]. Numerous investigations have indicated the feasibility of effectively eliminate immunotherapeutic resistance by promoting priming or activation of T lymphocytes, attracting or sustaining T cell-based immune response as well as favoring the immune-promoting the aforementioned TME, which is critical for cancer immunoediting and the component phases including elimination, equilibrium and escape [80, 81]. However, the developing of an effective T cell-based cytotherapy against cancer are extremely difficult largely due to the correspondence of tumor antigens to self MHC-associated fragments of self-proteins as well as the tumor heterogeneity [75, 82]. Encouragingly, functional cytolytic T lymphocyte (CTL)-mediated immune response has been reported with self-tumor antigen expression and efficient efficacy upon some cancer models and patients (e.g., melanoma, pancreatic carcinoma, breast cancer) [83, 84]. Collectively, T lymphocytes orchestrate various aspects of adaptive immunity, while subset delineation (e.g., TCF1+ progenitor subsets in exhausted CD8+ T cells, memory T cells) and tissue localization as well as targeted strategies (e.g., genetically engineered CAR-T or TCR-T) are key determinants of T lymphocyte function in immune responses [75, 77, 82, 85].

2.3.2 B lymphocytes

B cells, including the fetal liver-originated B-1 and bone marrow-derived B-2 subsets, are heterogeneous lymphocytes with antibody production and cytokine release capacity, which also represent a pivotal cellular constituent of humoral immunity and function critically in the maintenance of tolerance and immune regulation [86, 87, 88, 89]. Generally, B-1 lymphocytes, consist of B-1a and B-1b subsets, are part of innate immune system and mainly function in providing immunity to various specific pathogens via producing immunoglobulins [90]. B-2 lymphocytes, including the follicular B cells (FOB) and marginal zone B cells (MZB), have been considered as mediators of adaptive immunity and are capable of differentiating into memory cells [87].

Current studies have indicated the excessive inflammatory responses of regulatory B cells (Bregs) during autoimmune diseases, unresolved infections or chronic metabolic diseases, which also contribute to the dynamic balance of equilibrium required for tolerance by simultaneously reestablishing immune homeostasis and limiting ongoing immune responses [91, 92, 93]. Bregs encompassing all B cells for immune response suppression also play a promoting role in regulatory T cell (Treg) differentiation by accelerating anti-inflammatory cytokine (IL-10, IL-35, TGF-β) secretion and inhibiting proinflammatory cytokine production [91, 94]. Additionally, memory B cells (MBCs) have also been identified in humans and play an important role for the rapid development and response of protective immunity [95]. Collectively, with the aid of novel genetic and pharmacological technologies as well as the in-depth understanding of the phenotype, function and developmental processes, B cell-based cytotherapy has become a rapidly growing field in tumor immunotherapy [86, 94, 95, 96].

2.4 Engineered immunocytes

2.4.1 Chimeric antigen receptor-transduced T cells (CAR-Ts)

T lymphocytes with the antibody-like CAR structure expression are adequate to recognize the unique structures on the surface of tumor-associated cells or tumor cells, and in particular, the remarkable efficacy of CAR-Ts for the management of hematological malignancies [27, 82, 97]. For instance, we and other investigators have reported the remission of various hematopoietic malignancies such as refractory or relapsed B acute lymphoblastic leukemia (r/rB-ALL) and B-cell non-Hodgkin’s lymphoma (NHL) by utilizing the CAR-Ts targeting CD19, CD20, CD22 [98, 99, 100, 101, 102]. However, the adverse effects and the regulatory mechanisms of CAR-Ts-based tumor immunotherapy cannot be ignored [97, 103]. For example, Giavridis and the colleagues have reported the involvement of MΦ and IL-1 blockade in mediating and abating CAR-Ts-induced CRS [27, 104].

Despite the encouraging progress in hematologic malignancies, yet the applications of CAR-Ts-based cell therapy to solid tumors or as candidate pharmaceutical options have been challenging and suspicious [27, 68, 105, 106, 107]. In particular, the immunosuppressive TMEs of tumors represent the most important factor for limiting the efficacy of CAR-Ts-based immunotherapy [108]. It’s noteworthy that pioneering investigators have suggested the synergistic effect of oncolytic virus with CAR-Ts in improving the therapeutic effect of solid tumors [109]. For instance, Watanabe and the colleagues took advantage of the Mesothelin-redirected CAR-Ts (meso-CAR-Ts) and combined with an OAd-TNFa-IL2 oncolytic adenovirus for TNF-α and IL-2 expression, which increased CAR-Ts and host T cell infiltration to TME and thus showed enhanced efficacy upon human- pancreatic ductal adenocarcinoma (PDA)-xenograft immunodeficient mice [110].

2.4.2 T cell receptor-engineered T cells (TCR-Ts)

Besides chimeric antigen receptor (CAR) transduction, T lymphocytes can also be genetically engineered with αβ T cell receptor expression, which is capable of recognizing MHC-restricted peptide antigens and therefore poised to turn into an important pillar of cellular cancer immunotherapy [82, 111]. Generally, TCRs are capable of recognizing a relatively broad range of human leucocyte antigen (HLA)/specific peptide that can be expressed in the surface of patient’s T cells [112, 113, 114]. Therefore, TCR-Ts hold the potential to redirect the recognition of tumor-associated surface antigens and the removal of cancer cells. In details, TCR-Ts-based cytotherapy are principally intended to redirect circulating CD8+ T lymphocytes to the targets of expressing class I epitopes of HLA compared to those CD4+/CD8+ T cells with HLA Class-II epitopes. Collectively, TCR-transduced T cells hold promising prospective in targeting all intracellular proteins when peptide epitopes have been presented on the HLAs including over-expressed neoantigens and self -antigens, viral antigens [115]. Even though the characteristics of adaptive evolution in T cell immunity and the preferential expansion of T lymphocytes with high-affinity TCRs, yet whether the affinity maturation of T cells by clonal selection continues during the course of tumor development remains unresolved [114, 116].

2.4.3 CAR-transduced NK cells (CAR-NKs)

Compared to CAR-Ts, the genetically modified CAR-NKs not only inherit the splendid properties of adoptive NK cells including the aforementioned biological effects but also hold promising prospects for solid tumor administration without causing adverse effects including CRS, GvHD or immune cell- associated neurotoxicity syndrome (ICANS) [49, 50, 51, 117]. Thus, the CAR-NKs-based immunotherapy represents a virgin ground of immunotherapy innovation [118, 119, 120].

As to CAR transduction, a series of methodology has been developed to fulfill the high-efficient delivery demands via retrovirus-, lentivirus-, nonviral-mediated transfection with the range from 27–70% [121, 122]. In particular, the DNA transposon system composed of the sleeping beauty (SB) and the PiggyBac (PB) subsets is competent for delivering CAR structure into the genomes of induced pluripotent stem cells (iPSCs) or primary NK cells with high-efficient and long-lasting transgene expression [27, 123]. As to the CAR structures, a group of preclinical and clinical studies have reported the successful design and delivery of vectors carrying the cassettes of CAR-conjunct targets (e.g., CD19, CD5, CD137) into NK cells with the second- or third- or fourth-generation constructs against diverse malignancies, respectively [124, 125, 126, 127, 128]. Of note, Li et al announced the high-efficient generation of NK cells from human induced pluripotent stem cells (NK-CAR-iPSC-NKs) and superiority over T-CAR-expressing iPSC-NKs (T-CAR-iPSC-NKs), which highlighted the feasibility of the standardized and targeted CAR-NKs-based cancer immunotherapy in future [118].

2.4.4 CAR-transduced macrophages (CAR-ms)

State-of-the-art updates have indicated the feasibility of CAR-Ms for solid tumor management and virion load reduction [129, 130]. Differ from the other CAR-transduced immune cells, CAR-Ms are important sources of matrix metalloproteinase (MMPs), which can enter tumor tissue and degrade almost all extracellular matrix (ECM) components and thus destroy malignant tumor progression [130]. Strikingly, Klichinsky et al has reported the successful application of CAR-Ms in two solid tumor models and confirmed the prolonged overall survival and decreased tumor burden, which indicates the possibility of CAR-Ms-focused immunotherapeutic modalities [68, 131]. Notably, the therapeutic benefit of HER-2-targeting CAR-Ms was mediated by direct antigen-specific phagocytosis and indirect pro-inflammatory effects [132]. Recently, Zhang and the colleagues derived CAR-Ms from induced pluripotent stem cells (iPSC-CAR-Ms) with splendid properties such as cytokine secretion, polarization, enhanced phagocytosis and in vivo antitumor activity, which for the first time made iPSCs as unlimited source for “off-the-shelf” CAR-M generation [133].

Advertisement

3. Preclinical investigations and clinical applications

The current clinical and preclinical successes of cellular immunotherapy represent a remarkable point in cancer management, which also underscore the consequence of decoding the underlying tumor immunology [36, 134, 135]. Accumulating evidence has indicated the safety and effectiveness of cellular immunotherapy in recognizing and eliminating transformed cancer cells during various hematologic malignancies whereas those upon metastatic solid tumors are challenging and unsatisfactory [135, 136].

Generally, according to ClinicalTrials.gov (https://www.clinicaltrials.gov/) website, a total number of 4377 trials upon tumors by immunotherapy have been registered by the end of September, 2021. Of the aforementioned trials, there are 2463 in North America, 1089 in Europe and 834 in East Asia, respectively (Figure 1). As shown in Figure 1, among the indicated registered trials, there are 3944 interventional trials including 96 trials in early phase 1 stage, 957 in phase 1 stage, 776 in phase 1/2 stage, 1555 in phase 2 stage, 59 in phase 2/3 stage, 260 in phase 3 stage and 29 in phase 4 stage. In details, lymphoma (512 trials), leukemia (331 trials) and non-Hodgkin lymphoma (281 trials) are the top three indicates among hematologic malignancies, while lung neoplasms (822 trials), neuroectodermal tumors (751 trials) and digestive system neoplasms (696 trials) are the top three indicates among solid tumors (Figure 1). For instance, 26 trials of NK cell-based immunotherapy have been registered for a series of metastatic or recurrent tumor administration such as acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), juvenile myelomonocytic leukemia, liver cancer, breast cancer, non-small cell lung cancer (NSCLC), pancreatic cancer, ovarian cancer, cervical cancer, tongue cancer and esophageal cancer. Also, a number of 16 CAR-T-based immunotherapy (e.g., CD19, CD22, HER2, mesothelin, PSCA, MUC1, GPC3, BCMA, SLAMF7) has also been carried out for both hematological malignancies (e.g., relapsed/refractory multiple myeloma, acute lymphocytic leukemia and refractory indolent adult non-Hodgkin lymphoma) and solid tumors (e.g., advanced lung cancer, colon cancer, esophageal carcinoma, pancreatic cancer, prostate cancer, gastric cancer and hepatic carcinoma). For instance, we recently took advantage of the CD22, CD19–22 CAR-T therapy in patients with refractory or relapsed (r/r) B acute lymphoblastic leukemia (B-ALL) and confirmed the sustained remission after sequential treatment [98, 99, 100, 137, 138]. Additionally, other cancer immunotherapy has also been taken into practice including PD-1 (NCT02843204), TCR-T therapy (NCT03778814), DC-CIKs (NCT03190811, NCT01783951), Bevacizumab (NCT02857920).

Figure 1.

The overview of registered cancer immunotherapy worldwide.

As to preclinical investigations, immunotherapy has also acted as an “off-the-shelf” strategy and a promising candidate for clinical evaluation of cancer [21, 25, 102]. For example, Sommer et al reported the incorporation of allogeneic BCMA-CAR-Ts and CD20 mimotope-based intra-CAR off switch for conferring lymphodepletion resistance and reducing GvHD potential with the aid of the transcription activator-like effector nuclease (TALEN)-based gene editing. Notably, the CAR-T-based immunotherapy induced sustained antitumor responses and the cells maintained intrinsic phenotype and potency after scale-up manufacturing [139]. Recently, we reported the high-efficient generation of NK cells from peripheral blood with considerable killing activity upon K562 cells and summarized the latest updates upon allogeneic NK cell- and CAR-NK cell-based immunotherapy as well [21, 45]. Interestingly, we also found the delay of disease progression and improvement of leukemic hematopoietic microenvironment (LHME) in AML mouse models by blocking the migration of regulatory T cells (Treg) [140, 141].

Advertisement

4. Perspectives and future directions

For decades, comprehensive strategies for eliciting anticancer immunity have been extensively explored [34]. In particular, antitumor immunotherapies involving adoptive cellular transfer or immune checkpoint inhibitors are validated as effective and promising treatment option for hematopoietic malignancies and solid tumors [78]. Of the obstacles, tumor escape and cytotoxicity are the core burning issues in oncotherapy. To overcome the shortcomings, the systematic and precise understanding of TMEs and the relevant networks including pro- and anti-inflammatory cytokines, immunosuppressive cells, tumor-associated stroma, tumor hypoxia and metabolism as well as immune inhibitory checkpoints are collectively of great importance for improving the trafficking and delivery efficacy of CAR-transduced immune cells into the tumor site and helping solve the drawback of tumor antigen heterogeneity [108].

State-of-the-art updates have indicated the rosy and powerful implement of the anti-cancer immunotherapy including adoptive cellular transplantation and immune-checkpoint inhibitors for solid tumor and hematologic malignancy management in both preclinical studies and clinical practices [142, 143]. Nevertheless, the potentially acute and chronic adverse effects caused by immunotherapy-associated cytotoxicity have led to severe outcomes in tumor patients such as neurotoxicity, aGvHD, innate or acquired resistance, autoimmunity, nonspecific inflammation, cytokine storm syndrome (CSS), and the difficulty in realizing controllable modulation of the immune response, which are also the prerequisites and key challenges in the extensive implementation of immunotherapy for tumor, and in particular, the hurdles in adoptive T cell-based immunotherapy and double-edged properties of cancer immunoediting [75, 142, 143].

As to gene-edited adoptive immune cells, improvement strategies for enhancing the transfection efficiency and target selection as well as efficiently reducing the concomitant cytotoxicity during cancer immunotherapy are the key issue. For example, even though CAR-NKs exhibit inferior baseline cytotoxicity and preferable tumor killing activity when compared with other sources, yet the occasional issues should be systematically and thoroughly overcome by subsequent stimulation with optimized cytokine cock-tails [27, 144]. Therefore, it is of great importance to explore emerging features for the efficient development of novel immunotherapies, such as the selection of ideal CARs or TCRs targeting validated antigenic epitopes with well-characterized tumor cell expression and processing, enhancing immune cell effector function, persistence, expansion, trafficking, and memory formation by strategic selection of co-stimulated substrate cells, and novel technologies for gene-engineering [21, 27].

Of note, the variations and adverse effects in cancer immunotherapy reveals the heterogeneity and instability of the current immune cell products, and thus highlight the necessity and urgency of industrialization and standardization for clinical applications [145]. Generally, a cohort of core issues both in fundamental research and clinical practice of cancer immunotherapy need to be improved before large-scale applications [21, 146]. For instance, the screening criteria of healthy tissues for generating immune cell sources, the standardized regents and procedures for cell product preparation, the dose and frequency of cell transplantation, the delivery strategies and targets for engineered cells [147, 148, 149]. Therefore, it is of great importance for the generation of clinical-grade immune cell products based on good manufacturing practice (GMP) and convenient to universally improve life quality of patients with standard supervision. Additionally, multidisciplinary research has also highlighted the feasibility and prospective of nanomaterials (e.g., surface-conjugated nanoparticles, injectable scaffolds) as promising agents for cancer therapy attribute to the rapid progresses of nanobiotechnology and clinical biomedicine [18–20].

Collectively, comprehensive treatment strategies by combining the conventional remedies (eg., laparoscopic rectal surgery, robotic surgery, radiotherapy, chemotherapy, drugs), checkpoint blockade (e.g., PD-1/PD-L1, CTLA-4), vaccines (e.g., mRNAs), biomaterials (e.g., nanoparticles) with the aforementioned cellular immunotherapy will largely benefit the malignancy management and effectively reduce the cytotoxicity [21, 108, 150, 151, 152].

Advertisement

5. Conclusions

To date, hematological malignancies and metastatic solid tumors have caused a prevalence of over 10 million mortalities annually [3, 4, 5, 6]. Antitumor immunotherapy has served as a promising and alternative strategy for improving the outcomes of tumor patients as well as reducing the concomitant cytotoxicity. Objectively, there’s still a long way to go and a cohort of central issues need to be solved before large-scale application in cancer immunotherapy. Overall, cancer immunotherapy has become a notable and synergistic anti-tumor remedy for a variety of hematopoietic malignancies and locally advanced solid tumors.

Advertisement

Acknowledgments

The authors would like to thank the members in National Postdoctoral Research Station of Gansu Provincial Hospital, Hefei Institute of Physical Science, Chinese Academy of Sciences, and Institute of Health-Biotech, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd. for their technical support. We also thank the staff in Beijing Yunwei Biotechnology Development Co., LTD for their language editing service. This study was supported by grants from the project Youth Fund funded by Shandong Provincial Natural Science Foundation (ZR2020QC097), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019PT320005), Science and technology projects of Guizhou Province (QKH-J-ZK [2021]-107), the project Youth Fund funded by Jiangxi Provincial Natural Science Foundation (S2021QNJJL0277), Jiangxi Provincial Key New Product Incubation Program from Technical Innovation Guidance Program of Shangrao city (2020G002, 2020 K003), Natural Science Foundation of Tianjin (19JCQNJC12500), Jiangxi Provincial Novel Research and Development Institutions of Shangrao City (2020AB002, 2021F013).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

Not applicable.

Advertisement

Appendices and nomenclature

CAR-Tchimeric antigen receptor-transduced T cells
CAR-NKchimeric antigen receptor-transduced NK cells
CAR-Mchimeric antigen receptor-transduced macrophage
TCR-TT cell receptor-transduced T cells
LAKslymphokine-activated killer cells
TILstumor-infiltrating lymphocytes
CIKscytokine-Induced Killer cells
NKTnatural killer T
HCChepatocellular carcinoma
MDSCsmyeloid-derived suppressor cells
ADCCantibody-dependent cell-mediated cytotoxicity
ILCsinnate lymphoid cells
GvHDgraft-versus-host disease
HSCshematopoietic stem cells
CRScytokine release syndrome
CSCcancer stem cells
APCsantigen-presenting cells
SCLCsmall cell lung cancer
NSCLCnon-small cell lung cancer
DAMPsdamage-associated molecular patterns
macrophages
NOnitric oxide
CTLcytolytic T lymphocyte
NHLnon-Hodgkin’s lymphoma
ICANSimmune cell- associated neurotoxicity syndrome
iPSCsinduced pluripotent stem cells
MMPsmatrix metalloproteinase
ECMextracellular matrix
CMLchronic myelogenous leukemia
ALLacute lymphoblastic leukemia
TALENtranscription activator-like effector nuclease
LHMEleukemic hematopoietic microenvironment

References

  1. 1. Salamone JM, Lucas W, Brundage SB, Holloway JN, Stahl SM, Carbine NE, et al. Promoting scientist-advocate collaborations in cancer research: Why and how. Cancer Research. 2018;78(20):5723-5728
  2. 2. McEachron TA, Helman LJ. Recent advances in Pediatric cancer research. Cancer Research. 2021;81(23):5783-5799
  3. 3. Zaimy MA, Saffarzadeh N, Mohammadi A, Pourghadamyari H, Izadi P, Sarli A, et al. New methods in the diagnosis of cancer and gene therapy of cancer based on nanoparticles. Cancer Gene Therapy. 2017;24(6):233-243
  4. 4. Chilakamarthi U, Giribabu L. Photodynamic therapy: Past, present and future. Chemical Record. 2017;17(8):775-802
  5. 5. Wang Y, Liu T, Li X, Sheng H, Ma X, Hao L. Ferroptosis-inducing nanomedicine for cancer therapy. Frontiers in Pharmacology. 2021;12:735965
  6. 6. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA: a Cancer Journal for Clinicians. 2021;71(1):7-33
  7. 7. Bodey B, Siegel SE, Kaiser HE. Human cancer detection and immunotherapy with conjugated and non-conjugated monoclonal antibodies. Anticancer Research. 1996;16(2):661-674
  8. 8. Chen M, Hu S, Li Y, Jiang TT, Jin H, Feng L. Targeting nuclear acid-mediated immunity in cancer immune checkpoint inhibitor therapies. Signal Transduction and Targeted Therapy. 2020;5(1):270
  9. 9. Holyoake DLP, Smyth EC. Chemoradiotherapy or surgery for very early Esophageal squamous cancer: Can a nonrandomized trial give us the answer? Gastroenterology. 2021;161(6):1793-1795
  10. 10. Solaini L, Perna F, Cavaliere D, Vaccaro C, Avanzolini A, Cucchetti A, et al. Average treatment effect of robotic versus laparoscopic rectal surgery for rectal cancer. International Journal of Medical Robotics. 2021;17(2):e2210
  11. 11. Rallis KS, Lai Yau TH, Sideris M. Chemoradiotherapy in cancer treatment: Rationale and clinical applications. Anticancer Research. 2021;41(1):1-7
  12. 12. He Y, Liu QW, Liao HX, Xu WW. Microbiota in cancer chemoradiotherapy resistance. Clinical and Translational Medicine. 2021;11(1):e250
  13. 13. Conibear J, AstraZeneca UKL. Rationale for concurrent chemoradiotherapy for patients with stage III non-small-cell lung cancer. British Journal of Cancer. 2020;123(Suppl 1):10-17
  14. 14. Bordon Y. An RNA vaccine for advanced melanoma. Nature Reviews. Immunology. 2020;20(9):517
  15. 15. Sahin U, Oehm P, Derhovanessian E, Jabulowsky RA, Vormehr M, Gold M, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107-112
  16. 16. Rohatgi A, Kirkwood JM. Cancer vaccine induces potent T cell responses - but is it enough? Nature Reviews. Clinical Oncology. 2020;17(12):721-722
  17. 17. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Lower M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547(7662):222-226
  18. 18. Ning L, Zhu B, Gao T. Gold nanoparticles: Promising agent to improve the diagnosis and therapy of cancer. Current Drug Metabolism. 2017;18(11):1055-1067
  19. 19. Nazir S, Hussain T, Ayub A, Rashid U, MacRobert AJ. Nanomaterials in combating cancer: Therapeutic applications and developments. Nanomedicine. 2014;10(1):19-34
  20. 20. Liang P, Mao L, Dong Y, Zhao Z, Sun Q , Mazhar M, et al. Design and application of near-infrared nanomaterial-liposome hybrid Nanocarriers for cancer Photothermal therapy. Pharmaceutics. 2021;13(12):2070
  21. 21. Zhang L, Liu M, Yang S, Wang J, Feng X, Han Z. Natural killer cells: Of-the-shelf cytotherapy for cancer immunosurveillance. American Journal of Cancer Research. 2021;11(4):1770-1791
  22. 22. Daher M, Melo Garcia L, Li Y, Rezvani K. CAR-NK cells: The next wave of cellular therapy for cancer. Clinical & Translational Immunology. 2021;10(4):e1274
  23. 23. Yu SJ, Ma C, Heinrich B, Brown ZJ, Sandhu M, Zhang Q , et al. Targeting the crosstalk between cytokine-induced killer cells and myeloid-derived suppressor cells in hepatocellular carcinoma. Journal of Hepatology. 2019;70(3):449-457
  24. 24. Lu SJ, Feng Q. CAR-NK cells from engineered pluripotent stem cells: Off-the-shelf therapeutics for all patients. Stem Cells Translational Medicine. 2021;10(Suppl 2):S10-S17
  25. 25. Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. 'Off-the-shelf' allogeneic CAR T cells: Development and challenges. Nature Reviews. Drug Discovery. 2020;19(3):185-199
  26. 26. Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorganic Chemistry. 2021;112:104946
  27. 27. Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: From bench to bedside. Biomarker Research. 2022;10(1):12
  28. 28. Subauste CS, Dawson L, Remington JS. Human lymphokine-activated killer cells are cytotoxic against cells infected with toxoplasma gondii. The Journal of Experimental Medicine. 1992;176(6):1511-1519
  29. 29. Nakano K, Eura M, Chikamatsu K, Masuyama K, Ishikawa T. Characterization of transendothelial migratory lymphokine-activated killer cells. Japanese Journal of Cancer Research. 1996;87(4):395-400
  30. 30. Lafreniere R, Rosenberg SA. Successful immunotherapy of murine experimental hepatic metastases with lymphokine-activated killer cells and recombinant interleukin 2. Cancer Research. 1985;45(8):3735-3741
  31. 31. 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. The New England Journal of Medicine. 1985;313(23):1485-1492
  32. 32. 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. The New England Journal of Medicine. 1987;316(15):889-897
  33. 33. Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. Journal for Immunotherapy of Cancer. 2016;4:59
  34. 34. Badalamenti G, Fanale D, Incorvaia L, Barraco N, Listi A, Maragliano R, et al. Role of tumor-infiltrating lymphocytes in patients with solid tumors: Can a drop dig a stone? Cellular Immunology. 2019;343:103753
  35. 35. Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-infiltrating lymphocytes: Warriors fight against tumors powerfully. Biomedicine & Pharmacotherapy. 2020;132:110873
  36. 36. Yang Y. Cancer immunotherapy: Harnessing the immune system to battle cancer. The Journal of Clinical Investigation. 2015;125(9):3335-3337
  37. 37. Gao X, Mi Y, Guo N, Xu H, Xu L, Gou X, et al. Cytokine-induced killer cells As pharmacological tools for cancer immunotherapy. Frontiers in Immunology. 2017;8:774
  38. 38. Zhang Y, Ellinger J, Ritter M, Schmidt-Wolf IGH. Clinical studies applying cytokine-induced killer cells for the treatment of renal cell carcinoma. Cancers (Basel). 2020;12(9):2471
  39. 39. Giraudo L, Gammaitoni L, Cangemi M, Rotolo R, Aglietta M, Sangiolo D. Cytokine-induced killer cells as immunotherapy for solid tumors: Current evidence and perspectives. Immunotherapy. 2015;7(9):999-1010
  40. 40. Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, et al. Promising immunotherapy: Highlighting cytokine-induced killer cells. Journal of Cellular Biochemistry. 2019;120(6):8863-8883
  41. 41. Mohsenzadegan M, Peng RW, Roudi R. Dendritic cell/cytokine-induced killer cell-based immunotherapy in lung cancer: What we know and future landscape. Journal of Cellular Physiology. 2020;235(1):74-86
  42. 42. Bryceson YT, March ME, Ljunggren HG, Long EO. Activation, coactivation, and costimulation of resting human natural killer cells. Immunological Reviews. 2006;214:73-91
  43. 43. Xu J, Niu T. Natural killer cell-based immunotherapy for acute myeloid leukemia. Journal of Hematology & Oncology. 2020;13(1):167
  44. 44. Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer cells for cancer immunotherapy. Molecular Therapy. 2017;25(8):1769-1781
  45. 45. Liu M, Meng Y, Zhang L, Han Z, Feng X. High-efficient generation of natural killer cells from peripheral blood with preferable cell vitality and enhanced cytotoxicity by combination of IL-2, IL-15 and IL-18. Biochemical and Biophysical Research Communications. 2021;534:149-156
  46. 46. Cany J, van der Waart AB, Tordoir M, Franssen GM, Hangalapura BN, de Vries J, et al. Natural killer cells generated from cord blood hematopoietic progenitor cells efficiently target bone marrow-residing human leukemia cells in NOD/SCID/IL2Rg(null) mice. PLoS One. 2013;8(6):e64384
  47. 47. Kang L, Voskinarian-Berse V, Law E, Reddin T, Bhatia M, Hariri A, et al. Characterization and ex vivo expansion of human placenta-derived natural killer cells for cancer immunotherapy. Frontiers in Immunology. 2013;4:101
  48. 48. Hauswirth AW, Florian S, Printz D, Sotlar K, Krauth MT, Fritsch G, et al. Expression of the target receptor CD33 in CD34+/CD38-/CD123+ AML stem cells. European Journal of Clinical Investigation. 2007;37(1):73-82
  49. 49. Marofi F, Rahman HS, Thangavelu L, Dorofeev A, Bayas-Morejon F, Shirafkan N, et al. Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy. Stem Cell Research & Therapy. 2021;12(1):200
  50. 50. Hermanson DL, Kaufman DS. Utilizing chimeric antigen receptors to direct natural killer cell activity. Frontiers in Immunology. 2015;6:195
  51. 51. Geller MA, Cooley S, Judson PL, Ghebre R, Carson LF, Argenta PA, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011;13(1):98-107
  52. 52. Luna JI, Grossenbacher SK, Murphy WJ, Canter RJ. Targeting cancer stem cells with natural killer cell immunotherapy. Expert Opinion on Biological Therapy. 2017;17(3):313-324
  53. 53. Hu Y, Tian ZG, Zhang C. Chimeric antigen receptor (CAR)-transduced natural killer cells in tumor immunotherapy. Acta Pharmacologica Sinica. 2018;39(2):167-176
  54. 54. Kohli K, Pillarisetty VG. Dendritic cells in the tumor microenvironment. Advances in Experimental Medicine and Biology. 2020;1273:29-38
  55. 55. Tran Janco JM, Lamichhane P, Karyampudi L, Knutson KL. Tumor-infiltrating dendritic cells in cancer pathogenesis. Journal of Immunology. 2015;194(7):2985-2991
  56. 56. Gardner A, de Mingo PA, Ruffell B. Dendritic cells and their role in immunotherapy. Frontiers in Immunology. 2020;11:924
  57. 57. Waisman A, Lukas D, Clausen BE, Yogev N. Dendritic cells as gatekeepers of tolerance. Seminars in Immunopathology. 2017;39(2):153-163
  58. 58. Stevens D, Ingels J, Van Lint S, Vandekerckhove B, Vermaelen K. Dendritic cell-based immunotherapy in lung cancer. Frontiers in Immunology. 2020;11:620374
  59. 59. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nature Reviews. Cancer. 2012;12(4):265-277
  60. 60. Zhou J, Wang G, Chen Y, Wang H, Hua Y, Cai Z. Immunogenic cell death in cancer therapy: Present and emerging inducers. Journal of Cellular and Molecular Medicine. 2019;23(8):4854-4865
  61. 61. Zhang L, Yang X, Sun Z, Li J, Zhu H, Li J, et al. Dendritic cell vaccine and cytokine-induced killer cell therapy for the treatment of advanced non-small cell lung cancer. Oncology Letters. 2016;11(4):2605-2610
  62. 62. Arbour KC, Riely GJ. Systemic therapy for locally advanced and metastatic non-small cell lung cancer: A review. JAMA. 2019;322(8):764-774
  63. 63. Cavaillon JM. The historical milestones in the understanding of leukocyte biology initiated by Elie Metchnikoff. Journal of Leukocyte Biology. 2011;90(3):413-424
  64. 64. Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016;44(3):450-462
  65. 65. Luo Y, Shao L, Chang J, Feng W, Liu YL, Cottler-Fox MH, et al. M1 and M2 macrophages differentially regulate hematopoietic stem cell self-renewal and ex vivo expansion. Blood Advances. 2018;2(8):859-870
  66. 66. Zhang H, Li Z, Li W. M2 macrophages serve as critical executor of innate immunity in chronic allograft rejection. Frontiers in Immunology. 2021;12:648539
  67. 67. Goswami KK, Ghosh T, Ghosh S, Sarkar M, Bose A, Baral R. Tumor promoting role of anti-tumor macrophages in tumor microenvironment. Cellular Immunology. 2017;316:1-10
  68. 68. Anderson NR, Minutolo NG, Gill S, Klichinsky M. Macrophage-based approaches for cancer immunotherapy. Cancer Research. 2021;81(5):1201-1208
  69. 69. Mills CD, Lenz LL, Harris RA. A breakthrough: Macrophage-directed cancer immunotherapy. Cancer Research. 2016;76(3):513-516
  70. 70. Hibbs JB Jr, Vavrin Z, Taintor RR. L-arginine is required for expression of the activated macrophage effector mechanism causing selective metabolic inhibition in target cells. Journal of Immunology. 1987;138(2):550-565
  71. 71. Mills CD. Anatomy of a discovery: m1 and m2 macrophages. Frontiers in Immunology. 2015;6:212
  72. 72. Pei H, Qin J, Wang F, Tan B, Zhao Z, Peng Y, et al. Discovery of potent ureido tetrahydrocarbazole derivatives for cancer treatments through targeting tumor-associated macrophages. European Journal of Medicinal Chemistry. 2019;183:111741
  73. 73. Wang H, Tian T, Zhang J. Tumor associated macrophages (TAMs) in colorectal cancer (CRC): From mechanism to therapy and prognosis. International Journal of Molecular Sciences. 2021;22(16):8470
  74. 74. Baek SH, Lee HW, Gangadaran P, Oh JM, Zhu L, Rajendran RL, et al. Role of M2-like macrophages in the progression of ovarian cancer. Experimental Cell Research. 2020;395(2):112211
  75. 75. de Aquino MT, Malhotra A, Mishra MK, Shanker A. Challenges and future perspectives of T cell immunotherapy in cancer. Immunology Letters. 2015;166(2):117-133
  76. 76. Levite M. T cells plead for rejuvenation and amplification; with the Brain's neurotransmitters and neuropeptides we can make it happen. Frontiers in Immunology. 2021;12:617658
  77. 77. Kumar BV, Connors TJ, Farber DL. Human T cell development, localization, and function throughout life. Immunity. 2018;48(2):202-213
  78. 78. O'Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nature Reviews. Clinical Oncology. 2019;16(3):151-167
  79. 79. Seliger B, Massa C. Immune therapy resistance and immune escape of Tumors. Cancers (Basel). 2021;13(3):551
  80. 80. Smyth MJ, Ngiow SF, Ribas A, Teng MW. Combination cancer immunotherapies tailored to the tumour microenvironment. Nature Reviews. Clinical Oncology. 2016;13(3):143-158
  81. 81. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Current Opinion in Immunology. 2014;27:16-25
  82. 82. Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: Harnessing the T cell response. Nature Reviews. Immunology. 2012;12(4):269-281
  83. 83. Dudley ME, Rosenberg SA. Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nature Reviews. Cancer. 2003;3(9):666-675
  84. 84. Ryschich E, Notzel T, Hinz U, Autschbach F, Ferguson J, Simon I, et al. Control of T-cell-mediated immune response by HLA class I in human pancreatic carcinoma. Clinical Cancer Research. 2005;11(2 Pt 1):498-504
  85. 85. Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z, et al. Developmental relationships of four exhausted CD8(+) T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity. 2020;52(5):825-841 e828
  86. 86. Baba Y, Saito Y, Kotetsu Y. Heterogeneous subsets of B-lineage regulatory cells (Breg cells). International Immunology. 2020;32(3):155-162
  87. 87. Montecino-Rodriguez E, Dorshkind K. B-1 B cell development in the fetus and adult. Immunity. 2012;36(1):13-21
  88. 88. Nothelfer K, Sansonetti PJ, Phalipon A. Pathogen manipulation of B cells: The best defence is a good offence. Nature Reviews. Microbiology. 2015;13(3):173-184
  89. 89. Fillatreau S, Manfroi B, Dorner T. Toll-like receptor signalling in B cells during systemic lupus erythematosus. Nature Reviews Rheumatology. 2021;17(2):98-108
  90. 90. Wang Y, Liu J, Burrows PD, Wang JY. B cell development and maturation. Advances in Experimental Medicine and Biology. 2020;1254:1-22
  91. 91. Mauri C, Bosma A. Immune regulatory function of B cells. Annual Review of Immunology. 2012;30:221-241
  92. 92. Zhu Q , Rui K, Wang S, Tian J. Advances of regulatory B cells in autoimmune diseases. Frontiers in Immunology. 2021;12:592914
  93. 93. Catalan D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillon JC, et al. Immunosuppressive mechanisms of regulatory B cells. Frontiers in Immunology. 2021;12:611795
  94. 94. Akkaya M, Kwak K, Pierce SK. B cell memory: Building two walls of protection against pathogens. Nature Reviews. Immunology. 2020;20(4):229-238
  95. 95. Laidlaw BJ, Cyster JG. Transcriptional regulation of memory B cell differentiation. Nature Reviews. Immunology. 2021;21(4):209-220
  96. 96. Botia-Sanchez M, Alarcon-Riquelme ME, Galicia G. B cells and microbiota in autoimmunity. International Journal of Molecular Sciences. 2021;22(9):4846
  97. 97. Rodriguez-Garcia A, Palazon A, Noguera-Ortega E, Powell DJ Jr, Guedan S. CAR-T cells hit the tumor microenvironment: Strategies to overcome tumor escape. Frontiers in Immunology. 2020;11:1109
  98. 98. Pan J, Niu Q , Deng B, Liu S, Wu T, Gao Z, et al. CD22 CAR T-cell therapy in refractory or relapsed B acute lymphoblastic leukemia. Leukemia. 2019;33(12):2854-2866
  99. 99. Pan J, Zuo S, Deng B, Xu X, Li C, Zheng Q , et al. Sequential CD19-22 CAR T therapy induces sustained remission in children with r/r B-ALL. Blood. 2020;135(5):387-391
  100. 100. Pan J, Tan Y, Deng B, Tong C, Hua L, Ling Z, et al. Frequent occurrence of CD19-negative relapse after CD19 CAR T and consolidation therapy in 14 TP53-mutated r/r B-ALL children. Leukemia. 2020;34(12):3382-3387
  101. 101. Roselli E, Faramand R, Davila ML. Insight into next-generation CAR therapeutics: Designing CAR T cells to improve clinical outcomes. The Journal of Clinical Investigation. 2021;131(2):e142030
  102. 102. Frigault MJ, Maus MV. State of the art in CAR T cell therapy for CD19+ B cell malignancies. The Journal of Clinical Investigation. 2020;130(4):1586-1594
  103. 103. Hay KA. Cytokine release syndrome and neurotoxicity after CD19 chimeric antigen receptor-modified (CAR-) T cell therapy. British Journal of Haematology. 2018;183(3):364-374
  104. 104. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nature Medicine. 2018;24(6):731-738
  105. 105. 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. The New England Journal of Medicine. 2018;378(5):439-448
  106. 106. Kingwell K. CAR T therapies drive into new terrain. Nature Reviews. Drug Discovery. 2017;16(5):301-304
  107. 107. Hernandez-Lopez A, Tellez-Gonzalez MA, Mondragon-Teran P, Meneses-Acosta A. Chimeric antigen receptor-T cells: A pharmaceutical scope. Frontiers in Pharmacology. 2021;12:720692
  108. 108. Tahmasebi S, Elahi R, Esmaeilzadeh A. Solid Tumors challenges and new insights of CAR T cell engineering. Stem Cell Reviews and Reports. 2019;15(5):619-636
  109. 109. Wing A, Fajardo CA, Posey AD Jr, Shaw C, Da T, Young RM, et al. Improving CART-cell therapy of solid Tumors with oncolytic virus-driven production of a bispecific T-cell engager. Cancer Immunology Research. 2018;6(5):605-616
  110. 110. Watanabe K, Luo Y, Da T, Guedan S, Ruella M, Scholler J, et al. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight. 2018;3(7):e99573
  111. 111. Ecsedi M, McAfee MS, Chapuis AG. The anticancer potential of T cell receptor-engineered T cells. Trends Cancer. 2021;7(1):48-56
  112. 112. Attaf M, Roider J, Malik A, Rius Rafael C, Dolton G, Predergast AJ, et al. Cytomegalovirus-mediated T cell receptor repertoire perturbation is present in early life. Frontiers in Immunology. 2020;11:1587
  113. 113. Toya T, Taguchi A, Kitaura K, Misumi F, Nakajima Y, Otsuka Y, et al. T-cell receptor repertoire of cytomegalovirus-specific cytotoxic T-cells after allogeneic stem cell transplantation. Scientific Reports. 2020;10(1):22218
  114. 114. Schober K, Voit F, Grassmann S, Muller TR, Eggert J, Jarosch S, et al. Reverse TCR repertoire evolution toward dominant low-affinity clones during chronic CMV infection. Nature Immunology. 2020;21(4):434-441
  115. 115. Li D, Li X, Zhou WL, Huang Y, Liang X, Jiang L, et al. Genetically engineered T cells for cancer immunotherapy. Signal Transduction and Targeted Therapy. 2019;4:35
  116. 116. Redeker A, Arens R. ‘Reverse evolution’ in T cell biology. Nature Immunology. 2020;21(4):360-362
  117. 117. Berrien-Elliott MM, Cashen AF, Cubitt CC, Neal CC, Wong P, Wagner JA, et al. Multidimensional analyses of donor memory-like NK cells reveal new associations with response after adoptive immunotherapy for Leukemia. Cancer Discovery. 2020;10(12):1854-1871
  118. 118. 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 e185
  119. 119. Elahi R, Heidary AH, Hadiloo K, Esmaeilzadeh A. Chimeric antigen receptor-engineered natural killer (CAR NK) cells in cancer treatment; recent advances and future prospects. Stem Cell Reviews and Reports. 2021;17(6):2081-2106
  120. 120. Basar R, Daher M, Rezvani K. Next-generation cell therapies: The emerging role of CAR-NK cells. Blood Advances. 2020;4(22):5868-5876
  121. 121. Rostovskaya M, Fu J, Obst M, Baer I, Weidlich S, Wang H, et al. Transposon-mediated BAC transgenesis in human ES cells. Nucleic Acids Research. 2012;40(19):e150
  122. 122. Vargas JE, Chicaybam L, Stein RT, Tanuri A, Delgado-Canedo A, Bonamino MH. Retroviral vectors and transposons for stable gene therapy: Advances, current challenges and perspectives. Journal of Translational Medicine. 2016;14(1):288
  123. 123. Kim A, Pyykko I. Size matters: Versatile use of PiggyBac transposons as a genetic manipulation tool. Molecular and Cellular Biochemistry. 2011;354(1-2):301-309
  124. 124. Xu Y, Liu Q , Zhong M, Wang Z, Chen Z, Zhang Y, et al. 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. Journal of Hematology & Oncology. 2019;12(1):49
  125. 125. Chen KH, Wada M, Pinz KG, Liu H, Lin KW, Jares A, et al. Preclinical targeting of aggressive T-cell malignancies using anti-CD5 chimeric antigen receptor. Leukemia. 2017;31(10):2151-2160
  126. 126. Quintarelli C, Sivori S, Caruso S, Carlomagno S, Falco M, Boffa I, et al. Efficacy of third-party chimeric antigen receptor modified peripheral blood natural killer cells for adoptive cell therapy of B-cell precursor acute lymphoblastic leukemia. Leukemia. 2020;34(4):1102-1115
  127. 127. Xia J, Minamino S, Kuwabara K. CAR-expressing NK cells for cancer therapy: A new hope. Bioscience Trends. 2020;14(5):354-359
  128. 128. 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. 2021;137(5):624-636
  129. 129. Fu W, Lei C, Ma Z, Qian K, Li T, Zhao J, et al. CAR macrophages for SARS-CoV-2 immunotherapy. Frontiers in Immunology. 2021;12:669103
  130. 130. Zhang W, Liu L, Su H, Liu Q , Shen J, Dai H, et al. Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix. British Journal of Cancer. 2019;121(10):837-845
  131. 131. Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nature Biotechnology. 2020;38(8):947-953
  132. 132. Mukhopadhyay M. Macrophages enter CAR immunotherapy. Nature Methods. 2020;17(6):561
  133. 133. Zhang L, Tian L, Dai X, Yu H, Wang J, Lei A, et al. Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions. Journal of Hematology & Oncology. 2020;13(1):153
  134. 134. Wang W, Jiang J, Wu C. CAR-NK for tumor immunotherapy: Clinical transformation and future prospects. Cancer Letters. 2020;472:175-180
  135. 135. Morgan MA, Buning H, Sauer M, Schambach A. Use of cell and genome modification technologies to generate improved “off-the-shelf” CAR T and CAR NK cells. Frontiers in Immunology. 1965;2020:11
  136. 136. 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
  137. 137. Pan J, Deng B, Ling Z, Song W, Xu J, Duan J, et al. Ruxolitinib mitigates steroid-refractory CRS during CAR T therapy. Journal of Cellular and Molecular Medicine. 2021;25(2):1089-1099
  138. 138. Tan Y, Pan J, Deng B, Ling Z, Song W, Xu J, et al. Toxicity and effectiveness of CD19 CAR T therapy in children with high-burden central nervous system refractory B-ALL. Cancer Immunology, Immunotherapy. 2021;70(7):1979-1993
  139. 139. Sommer C, Boldajipour B, Kuo TC, Bentley T, Sutton J, Chen A, et al. Preclinical evaluation of allogeneic CAR T cells targeting BCMA for the treatment of multiple myeloma. Molecular Therapy. 2019;27(6):1126-1138
  140. 140. Wang R, Feng W, Wang H, Wang L, Yang X, Yang F, et al. Blocking migration of regulatory T cells to leukemic hematopoietic microenvironment delays disease progression in mouse leukemia model. Cancer Letters. 2020;469:151-161
  141. 141. Ren J, Han L, Tang J, Liu Y, Deng X, Liu Q , et al. Foxp1 is critical for the maintenance of regulatory T-cell homeostasis and suppressive function. PLoS Biology. 2019;17(5):e3000270
  142. 142. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nature Reviews. Drug Discovery. 2019;18(3):175-196
  143. 143. Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA: a Cancer Journal for Clinicians. 2020;70(2):86-104
  144. 144. Balassa K, Rocha V. Anticancer cellular immunotherapies derived from umbilical cord blood. Expert Opinion on Biological Therapy. 2018;18(2):121-134
  145. 145. Koepsell SA, Miller JS, McKenna DH Jr. Natural killer cells: A review of manufacturing and clinical utility. Transfusion. 2013;53(2):404-410
  146. 146. Pahl JHW, Koch J, Gotz JJ, Arnold A, Reusch U, Gantke T, et al. CD16A activation of NK cells promotes NK cell proliferation and memory-like cytotoxicity against cancer cells. Cancer Immunology Research. 2018;6(5):517-527
  147. 147. Lu C, Guo C, Chen H, Zhang H, Zhi L, Lv T, et al. A novel chimeric PD1-NKG2D-41BB receptor enhances antitumor activity of NK92 cells against human lung cancer H1299 cells by triggering pyroptosis. Molecular Immunology. 2020;122:200-206
  148. 148. Wrona E, Borowiec M, Potemski P. CAR-NK cells in the treatment of solid Tumors. International Journal of Molecular Sciences. 2021;22(11):5899
  149. 149. Jayaraman J, Mellody MP, Hou AJ, Desai RP, Fung AW, Pham AHT, et al. CAR-T design: Elements and their synergistic function. eBioMedicine. 2020;58:102931
  150. 150. Grosser R, Cherkassky L, Chintala N, Adusumilli PS. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid Tumors. Cancer Cell. 2019;36(5):471-482
  151. 151. Zhang F, Stephan SB, Ene CI, Smith TT, Holland EC, Stephan MT. Nanoparticles that reshape the tumor milieu create a therapeutic window for effective T-cell therapy in solid malignancies. Cancer Research. 2018;78(13):3718-3730
  152. 152. Yong SB, Chung JY, Song Y, Kim J, Ra S, Kim YH. Non-viral nano-immunotherapeutics targeting tumor microenvironmental immune cells. Biomaterials. 2019;219:119401

Written By

Leisheng Zhang and Hui Cai

Submitted: 25 March 2022 Reviewed: 05 May 2022 Published: 12 June 2022