Open Access

Immunotherapy: A new target for cancer cure (Review)

  • Authors:
    • Fuqian Zhao
    • Daofu Shen
    • Mingquan Shang
    • Hao Yu
    • Xiaoxue Zuo
    • Lixin Chen
    • Zhijin Huang
    • Lianxing Li
    • Lei Wang
  • View Affiliations

  • Published online on: March 28, 2023     https://doi.org/10.3892/or.2023.8537
  • Article Number: 100
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer is the leading cause of death globally and there is a worldwide cancer epidemic. Immunotherapy has emerged as a promising anticancer therapy. In particular, oncolytic viruses destroy cancer cells without destroying normal tissue via viral self‑replication and anti‑tumor immune responses, showing potential for cancer therapy. The present review discusses the role of the immune system in the treatment of tumor. The strategies for treating tumors are briefly introduced from aspects of active immunization and passive immunotherapy and the dendritic cell vaccines and oncolytic viruses are highlighted, as well as use of blood group A antigen in the treatment of solid tumors.

Introduction

Cancer is the main cause of death in the world and there is even a view that the world is experiencing a cancer epidemic (1). In 2018, China had 4.3 million new cancer cases and 2.9 million cancer deaths (2). The huge economic burden puts notable pressure on the medical system and patient wellbeing. Surgical treatment is considered to be the most effective way to treat tumors; after the advent of chemotherapy in 1940 and targeted therapy in the late 1990s, immunotherapy is the third important era of cancer treatment (3). In the past few decades, anti-cancer immunotherapy has transformed from an emerging tumor treatment theory to a well-known alternative tumor therapy. Anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4 and anti-programmed death receptor 1 (PD)-1/PD-ligand (L) 1, as immunotherapy, for the treatment of melanoma, colorectal cancer and breast cancer plays an important role (46).

Immunotherapy refers to the method of artificially enhancing or inhibiting the immune function of the body to cure diseases. Tumor immunotherapy is based on the immune surveillance theory proposed by Frank Macfarlane Burnet and Lewis Thomas (7). The theory of immune surveillance posits that the system can play a surveillance role to identify and eliminate foreign components or mutant cells that express new antigens to maintain the stability of the host environment. When the immune function is low and cannot effectively eliminate foreign or mutated cells, tumors may occur (8). Dunn et al (9) put forward the theory of immune editing, which further improved the framework of tumor immunity. The immunoediting theory posits that the development of tumors needs to go through three stages: Immune clearance, balance and escape (10). Tumor cells that can escape the immune system may survive natural selection. If tumors are regarded as immunogens, decades of research have not found valuable tumor antigen-regulated immune escape theory, and a large number of experiments have proved that tumor stem cells with reduced expression of tumor antigens further prove that the search for tumor-specific antigens or wrong research direction (11,12). Hypoxia at the tumor site may cause attenuation of tumor immunogens (13). It can hypothesized that the immune system recognizes and destroys tumor cells expressing strong immunogenicity, while tumor cells with weak (or no) immunogenicity selectively survive and eventually form tumors. Immunotherapy may become the most advantageous tool to overcome this (14). The relationship between the immune system and tumors is complicated. In 1891, American doctor William Coley discovered that postoperative infection of Streptococcus pyogenes in patients with sarcoma could cause tumor regression. This discovery provided a new idea for cancer immunotherapy (15). With the emergence of new technologies such as humanized antibodies, virus packaging and gene high-throughput sequencing, tumor therapy has achieved rapid development. This review summarizes strategies for immunotherapy to treat cancer (16,17).

Association between the immune system and tumor

The immune system consists of immune organs (bone marrow, thymus, spleen, lymph nodes, tonsils, small intestinal Peyer's lymph nodes, appendix, thymus, etc.), immune cells (lymphocytes, mononuclear phagocytes, neutrophils, basophils, eosinophils, etc.) Granulocytes, mast cells, platelets, etc.), and immune molecules (complement, immunoglobulin, interferon, interleukin, tumor necrosis factor and other cytokines, etc.) (18). The immune system recognizes and eliminates antigenic foreign bodies, coordinating with other systems of the body, and maintaining the stability of the host environment and physiological balance (19). Immune organs are be divided into central (bone marrow and thymus) and peripheral immune organs (spleen, lymph nodes and tonsils); immune cells occur, differentiate and mature in central organs and B lymphocytes colonize and proliferate in peripheral organs, where the immune response primarily occurs (20,21). Immune cells comprise innate (dendritic (D) and natural killer (NK) cells and macrophages) and adaptive immune cells (T and B cells) (22). Immune molecules comprise membrane-type (such as T and B cell receptor (CR), adhesion and major histocompatibility complex (MHC) molecules and cytokine receptors) and secreted molecules (such as immunoglobulin, complement and cytokines) (23,24). The most important function of the innate immune system is to respond quickly to infection or inflammation and to recruit innate immune cells or activate complement via cytokines secreted by the injured site (such as ILs and chemokines) (25). Both B and T cells originate from a common lymphoid progenitor cell (B cells mature in the bone marrow and T cells mature in the thymus) and mediate humoral and cellular immunity, respectively (26). B cells participate in production of antibodies, and T cells participate in proliferation of B cells, directly attack pathogens and regulate immune responses (27). Adaptive immunity is associated with immune memory and long-term effects of the immune system and serves a key role in fighting tumors (28).

The tumor antigen-specific T cells produced by adaptive immunity are considered to be the key factor in killing tumors (29). This process is inseparable from the innate immune response and includes the following steps: Phagocytes engulf and digest tumor cells to produce tumor antigens; antigen-presenting cell (APC) cross-presentation of tumor antigens to T cells; initiation and activation of initial T cells; transport and infiltration of activated T cells into the tumor microenvironment (TME) and activated CTL-mediated malignant cell death. The generation of tumor antigen-specific T cells reflects the coordination between the innate and the adaptive immune system (3033). This dynamic interaction is guided by the phenotype and function of innate immune cells to influence tumor antigen-specific T cells, resulting in different biological states (tolerance or responsiveness) (34). As the APC in TME, DCs initiate cancer immunity by cross-presenting tumor-associated antigens to naive T cells (35). Although antigen-loaded DCs are potent stimulators of T cell activation, DCs activate antigen-specific CTL expansion through the CD40/CD40L pathway (36). CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily that is expressed in large quantities on the DC membrane. After recognizing its homologous antigen, CD4+ T helper (Th) cells can express CD40L and then combine with the complementary CD40. After activation, the expression of MHC II, CD80and CD86 on the surface of DCs is increased, which supports T cell activation (37). Conventional DCs (cDCs) in mice are divided into two lineages with different functions: CD103+ cDC1 lineage is responsible for the initiation of CD8+ CTL and CD11b+ cDC2 lineage is associated with priming CD4+ Th cells (38,39). In a melanoma mouse model, CD103+ cDC1s promote T cell recruitment to the TME by releasing the chemokine CXCL9/10 (40). Similar to DCs, macrophages are key innate immune cells that promote or hinder the activation of effector T cells (41). This is because their functional characteristics are affected by signals from the surrounding microenvironment and the cell phenotype has strong plasticity. IFN-γ and toll-like receptor agonists induce differentiation into M1 phenotypes related to anti-tumor activity; IL-4 and IL-13 induce differentiation into M2, which is associated with tumor-promoting activity (42,43). M1 macrophages are mainly involved in the immune response against foreign pathogens and M2 macrophages help wound healing and secrete anti-inflammatory cytokines (44). In tumorigenesis and metastasis, M1 macrophages serve an adaptive immune surveillance function, while M2 macrophages inhibit the anti-tumor immune function of T cells (45). M1-like macrophages in TME phagocytose tumor cells and present tumor antigens to initiate the anti-tumor activity of CD8+ T cells (46). Macrophages residing in tissues other than tumors can also transfer phagocytosed tumor antigens to DCs to trigger an adaptive immune response by inducing CTL cross-reactions (47). Macrophages are commonly used by tumors to suppress adaptive immune responses. M2 macrophages inhibit T cell activation by secreting IL-10 to destroy the TCR (48). In addition, tumor-infiltrating macrophages express a variety of immune checkpoint proteins (such as PDL1) that bind to T cell inhibitory signal receptors to inhibit cell function (49). CD169+ macrophages capture tumor antigens to prevent them inducing an immune response (Fig. 1).

Passive immunotherapy

Anti-cancer immunotherapy is classified as passive or active according to the ability to activate the host immune system against malignant cells. Tumor-targeted monoclonal antibodies and adoptively transferred T cells are considered passive forms of immunotherapy because exhibit inherent anti-tumor activity (50). Anti-cancer vaccines and immune checkpoint inhibitors that only exert anti-cancer effects when the host immune system is involved are classic examples of active immunotherapy (51).

Immunoglobulins, also called antibodies, are the first molecules involved in specific immune responses (52). Antibodies with unique specificities that recognize different target molecules have been used to attack tumor cells that express certain antigens (53). There are five mechanisms for tumor-targeting antibodies to produce anti-tumor effects. Therapeutic antibodies, such as epidermal growth factor receptor (EGFR)-specific monoclonal antibody cetuximab for the treatment of head and neck and colorectal cancer, inhibit the signaling pathways required for tumor cell survival or progression. Therapeutic antibodies, such as tigatuzumab, a monoclonal antibody specific to TNF receptor superfamily member 10B, activate potentially lethal receptors expressed on the surface of tumor cells. Immunoconjugates (tumor antigen-specific antibodies conjugated to toxins or radionuclides), such as gemtuzumab and ozogamicin (an anti-tumor cell membrane-expressed CD33 calicheamicin conjugate approved for use in patients with acute myeloid leukemia), directly kill tumor cells. Simple antibodies directed against tumor-specific or -associated antigens (such as rituximab, which is currently approved for the treatment of chronic lymphocytic leukemia and non-Hodgkin's lymphoma) work by activating antibody-dependent cell-mediated cytotoxicity and cellular phagocytosis and complement-dependent cytotoxicity. Bispecific T cell conjugates are composed of two monoclonal antibodies from different monoclonal antibodies (5459). An artificially modified antibody, the chimeric protein of its variable region, one targets tumor cells, and the other specifically targets T cell surface antigens, shortening the distance between T cells and tumor cells in space, allowing T cells to directly Kill tumor cells. For example, blinatumomab, a therapeutic antibody that targets the CD19 molecule on tumor cells and the CD3 molecule on T cells, is used in the treatment of Philadelphia chromosome-negative precursor B cell acute lymphoblastic leukemia (60). Approved antibodies targeting tumor cells (such as Catumaxoma) and other antibody drugs (such as Veltuzumab) under development belong to the IgG class. IgA molecules are also used as anticancer agents. For example, the anti-EGFR IgA2 containing the variable region of cetuximab significantly decreases the number of metastases in a melanoma cell lung metastasis model of transgenic mice expressing human EGFR (61). This effect of IgA2 lasts a week longer than the corresponding IgG cetuximab (62,63). IgE is another antibody class being explored as a potential cancer treatment (64). The research on the function of using IgE-mediated immune response against tumor cells in the context of cancer belongs to the rapidly developing allergoncology field (Fig. 2) (65).

Cell therapy refers to the transfer of autologous or allogeneic cell material into the body for medical purposes (66). Adoptive cell transfer is a cell-based anti-cancer immunotherapy. The usual practice is to use immune enhancers to activate blood circulation or tumor-infiltrating lymphocytes to achieve the purpose of fighting tumors (67). Other anti-cancer immunotherapies involve live cell transfusion, such as hematopoietic stem cell transplantation, to rebuild a healthy, allogeneic immune system; adoptive cell transfer is the infusion therapy of immune cells with potential antitumor immune activity (68). Interventions based on DCs are different from the aforementioned cell therapies. Infused DCs do not have anti-cancer activity but can be used as anti-cancer vaccines to trigger tumor-targeted immune responses (69). The cellular immune response against tumors primarily depends on T cells. A large number of antigens have been identified in tumors that are recognized by T cells, suggesting the potential role of T lymphocytes in anti-tumor immune responses (70). In certain patients with melanoma or pancreatic cancer, Epstein-bar virus-associated malignancy and murine tumor models, functional CTL have been shown to fight tumor cells that express tumor antigens (71,72). The permanent establishment of memory immune T cells serves a key role in preventing tumor recurrence (73). However, due to the poor immunogenicity of most tumors, it is difficult to cultivate a lymphocyte population with sufficient affinity for TCRs, and it is difficult to introduce engineered surface receptors with enhanced affinity for a tumor-specific antigen (74). Chimeric antigen receptors (CARs) consist of antibody-derived antigen recognition domains that are connected to the internal T cell signaling domain and recognize antigen targets through a mechanism different from that of classical TCR (75). Unlike traditional TCRs that recognize intracellular peptide antigens presented by MHC molecules, CARs directly recognize antigens expressed on the surface of tumor cells so are not limited by the patient HLA subtype, and can recognize a variety of antigen structures, including proteins, carbohydrates and glycolipids (60). Gene therapy viral vectors can transfect genes encoding CAR constructs into T cells to express high-affinity extracellular antigen-recognition moieties and membrane proteins derived from monoclonal antibody single-chain variable fragment-binding TCR signaling domains (76). The internal domain of CAR is originally derived from the CD3ζ chain of traditional TCR, and after technical development, it can include one or more costimulatory domains (most commonly CD28 and 41BB) to enhance the persistence and cytotoxicity of CAR-expressing cells (77).

Active immunotherapy

DC cells were first discovered in 1868 by Langerhans in the skin (78). In 1973, Ralph Steinman discovered similar cells in the spleen of mice and proposed dendrites and their functions (79). In March 2007, after Steinman was diagnosed with advanced pancreatic cancer, he used his own developed DC cells for treatment, extending his life expectancy from a few months to 4.5 years (80). American Food and Drug Administration (FDA)-approved Sipuleucel-T is a vaccine against advanced castration-resistant prostate cancer and the first therapeutic DC vaccine against cancer. DCs are the key cells that initiate and respond to pathogens to activate naive T cells (81). DC cells also serve a key role in maintaining immune tolerance. DCs are derived from B cells, macrophages, Langerhans cells and inflammatory/monocytes Together with DCs, B cells and macrophages, are considered to be antigen-presenting cells (82). Pattern-recognition receptor (PRR) ligand expressed on DCs can lead to activation and can induce simultaneous antigen uptake and processing to produce peptide antigen, together with MHC molecules, pMHC complexes are formed (83). In secondary lymphoid organs, DCs present pMHC complexes to naive T cells, presents pMHC to TCRs by activating or inhibiting membrane receptors. Subsequently, soluble cytokines are expressed and secreted by DCs as signaling molecules (84). Although they are ubiquitous in most tissue, the absolute number of DCs is very low. For example, mature DCs only account for ~1% of the total peripheral blood mononuclear cells (85). With the improvement of DC in vitro derivation, a variety of precursor cells can be used to prepare DCs, such as non-proliferative CD14+ monocytes from peripheral blood and proliferative CD34+ precursor cells from bone marrow and blood in the umbilical cord (86). In addition, by direct transdifferentiation or indirect dedifferentiation and differentiation, myeloid and lymphoid DCs can differentiate to DCs (87).

There are several forms of DC-based immunotherapies, most of which involve isolation of circulating monocytes from patients or donors and their expansion and differentiation in vitro to promote the maturation of DCs by cytokines (such as TNF-α, IL-1β) (78). Immature DCs have an immunosuppressive function rather than an immune enhancing function, and the use of macrophage colony-stimulating factor to stimulate immature DCs to differentiate into mature DCs plays an important role in their anti-tumor function (88).

For tumor cells with low expression of MHC I, their tumor antigen presentation ability is weakened and it is difficult to activate T cells to kill tumor cells (89). To better present tumor antigens to T cells, DC vaccines introduce tumor-associated antigens (including proteins, peptides or tumor lysate) from patients into DCs. The pMHC is expressed on the cell surface to initiate an immune response. The preparation methods of DC vaccines include directly sensitizing DCs with tumor antigens and cell lysates to produce activity; viral vectors encoding tumor associated antigen (TAA) gene to infect DCs to express the corresponding antigens and Tumor antigen mRNA is electroporated or chemically transfected into DC cells or fused with DC cells using a fusion agent with tumor cells expressing tumor antigens (Fig. 3) (90,91).

Sensitization of DCs with tumor antigens and cell lysates is the most common method for preparing DC vaccines (92). Immature DCs phagocytose tumor antigens and differentiate into mature DCs in vitro. These cells carrying antigenic information are returned to the body to activate the anti-tumor immune response (93). After DCs carrying antigens, such as carcinoembryonic antigen and melanoma antigen-A1, are returned to patients with lung cancer, the body can produce specific T lymphocytes (94). However, tumor-associated antigens are not unique to tumor cells and it is difficult to induce a specific immune response against tumor cells. Therefore, sequencing and mass spectrometry have been used to analyze and identify neoantigens on the surface of tumor cells (95). Excised tumor tissue is lysed by ultrasonic disruption, subjected to repeated freezing and thawing and used as an active ingredient to prepare DCs that induce anti-tumor responses (96). DCVax®-L is a personalized DC vaccine sensitized by lysate of malignant glioblastoma developed by Northwest Biotherapeutics in the United Kingdom that infects DCs with viral vectors to insert the gene encoding TAA into a lentivirus, recombinant poxvirus or adenovirus vector (97). After the virus infects DCs, it expresses TAA and maturation is induced. The low efficiency of virus infecting DC means the development of DC vaccines has been limited (98). Tumor cells and DCs are fused under the action of fusion agents, and the fused cells not only have the function of DCs, but also express tumor antigens on the cells.

DC vaccines are highly immunogenic and highly specific. Electrical, viral and chemical fusion are commonly used methods for preparing DC vaccines. Due to the instability of electric fusion, this technology is no longer used (99). Viruses commonly used to induce animal cell fusion include Sendai, Newcastle disease and herpes virus (100,101). Inactivated Sendai virus is used to induce cell fusion; the fusion rate is high and it is suitable for various types of animal cell. Because Sendai virus is unstable, the preparation process it is cumbersome and may affect the normal function after entering the cell (102). Polyethylene glycol has good water solubility and adhesion, and is a commonly used chemical reagent for the fusion of cells. In DC immunotherapy, the immunosuppressive nature of the tumor microenvironment inhibits the antigen presentation ability of DC cells (103), the limited ability of DC cells to target tumors cannot specifically recognize tumor antigens, and the specific T cells produced by DCs after presenting antigens Cells have low affinity for tumor cells (35,93,104). Due to the multiple roles of DCs in the immune response, DC vaccines are still a promising treatment.

Cytokines are key biomolecules that communicate with each other and exert biological functions in immune cells (105). As a family of proteins, they regulate almost all biological functions of cells via autocrine, paracrine or endocrine effects. Based on the powerful immunomodulatory ability, the immunotherapy of cancer with cytokines has been tried (106,107). The therapeutic effect of IFN is considered important in cancer immunoediting and has been studied in many clinical trials (108,109). Type I IFN and IL-2 have been approved by the US FDA for the treatment of certain types of malignancy, such as Melanoma, metastatic renal cell carcinoma (110,111). Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been approved for adjuvant therapy of malignant tumors due to its ability to stimulate proliferation and differentiation of immune cells (112). In addition, GM-CSF promotes antigen presentation by DCs, making it widely used in tumor vaccines (113,114). Although the antitumor activity of cytokines has been observed in many studies (115117), few cytokines induce complete tumor regression. To the best of our knowledge, the mechanism of action of immune-stimulating cytokines has not been fully explored and some tumor treatment modalities in the clinic may promote cytokine cascades with unexplained potentially lethal effects (118). Radiotherapy and chemotherapy trigger a cytokine storm in the tumor stroma, including release of the pro-tumor cytokines IL-6 and TNFα. Apoptotic tumor cells activate macrophages to produce pro-inflammatory mediators and cellular debris can also stimulate anti-tumor immunity; therefore, dead and dying tumor cells contribute to a TME that may promote tumor progression (119,120). The anti-tumor function of cytokines is complex, and studies have shown that their anti-tumor activity depends on the host immune system (121123). Cytokines are functionally divided into pro- and anti-inflammatory. Pro-inflammatory cytokines (such as IL-1α/β, TNF-α/β, IL-6, IL-11, IL-18 and IFN-γ) upregulate inflammatory responses and enhance recruitment, infiltration and resistance of immune cells to tumor site (124126). Anti-inflammatory cytokines (such as IL-10, IL-6, TGF-β, IL-27 and IL-35) downregulate inflammatory responses and promote tissue healing and tumor growth (127,128). Cytokine-induced inflammatory responses are context-dependent; the same cytokines induce pro-inflammatory or anti-inflammatory responses depending on factors such as target cells, dose, and presence of other cytokines (129). Cytokine classification in tumor therapy stems from the association between cytokines and T cell responses. T cells differentiate into cell populations with different functions, characterized by production of certain cytokine groups. Th1 cells produce type 1 cytokines such as IL-2, IL-12 and IFN-γ; Th2 cells produce type 2 cytokines, such as IL-4, IL-5, IL-6, IL-10 and IL-13; regulatory T cells produce IL-10 and TGF-β (130). In general, type 1 cytokines mediate the development of strong cellular immune responses, while type 2 cytokines facilitate strong humoral immune responses (131). Tumors are often associated with a tolerant and immunosuppressive microenvironment. Cytokine-mediated therapy uses type 1 cytokines to stimulate anticancer immune responses (132). Vaccine-based therapies use type 2 cytokines as adjuvants based on their role in B cell maturation, while autoimmune diseases may benefit from regulatory cytokines (133). In many cases, these distinctions are not sufficient to classify cytokines because their effects on the immune system are complex. For example, IL-18 can promote Th2-biased cytokine production by T cells but in the presence of IL-15 or IL-12, IL-18, leads to potent Th1-biased cytokine production (134). Furthermore, type 1 cytokines are not limited to cellular immune responses, as they contribute to the development of certain antibody classes and functional differentiation of B cells (135).

Cytokine storm involves a variety of cytokines such as TNF-α, IL-1, IL-6, IL-12, IFN-α, IFN-β, IFN-γ, monocyte chemotactic protein 1 and IL-2 (136). The phenomenon of rapid and massive production of IL-8 is an important cause of acute respiratory distress syndrome and multiple organ failure (137). Injected CAR T cells to treat CD19+ lymphoma induce a cytokine storm, with levels of IFN-γ and IL-6 exceeding physiological levels, due to high levels of activated CAR T cells (138). The cytokines that mediate the cytokine storm are achieved not by CAR T cells but by macrophages, and their damage to the body can be mitigated by the use of IL-6 and IL-1 drugs (139). Glucocorticoids and IL-6 inhibitors are also effective in treating this type of cytokine storm (140). To the best of our knowledge, no cases of cytokine storm or elevated IL-6 levels have been reported in NK cell CAR therapy.

In conclusion, cytokines are potent but complex immune mediators. Developing cytokine drugs is a challenge that requires a deep understanding of cytokine biology and contemporary biotechnology to exploit their antitumor activity, while minimizing toxicity. In future, how to confine the action of cytokines to the desired site to avoid systemic pro-inflammatory effects and how to incorporate these treatments into combination immunotherapy strategies should be investigated.

Future immunotherapy

Oncolytic viruses refer to non-pathogenic viruses that specifically infect tumor cells and cause their death. Oncolytic viruses are an emerging class of antitumor immunotherapy (141). The effectiveness of oncolytic viruses depends on sufficient numbers of oncolytic virus to infect tumor cells. Because oncolytic viruses have intrinsic anticancer activity, they are considered passive immunotherapy (142). Although not fully understood, it is hypothesized that oncolytic viruses mediate anti-tumor activity through two distinct mechanisms of action: Selectively replicating within tumor cells, resulting in direct lytic effects or inducing systemic antitumor immune response (143). Specifically, oncolytic virus therapy relies on tumor cell-specific changes associated with tumor characteristics, including increased receptor expression, impaired antiviral response and alterations in cellular metabolism; it is hypothesized that oncolytic virus replication is limited to the tumor site and healthy tissue is not harmed (144). In addition to directly lysing tumors, oncolytic viruses induce extracellular matrix remodeling, thus exerting anti-angiogenic effects (145). In the anti-tumor immune response, tumor cells release cytoplasmic components such as intracellular tumor-associated antigens, damage and pathogen-associated molecules after death to stimulate the body's innate immunity, and a large number of cytokines and chemokines are produced to promote subsequent specific immunity of the tumor. Mediate the maturation of APC and enhance its antigen presentation ability, promote the initiation, activation, proliferation, transport, memory formation, cytokine release and cytotoxic activity of polyclonal T cells, and generate systemic anti-tumor immune response (146). Various oncolytic viruses have been used to treat different forms of cancer, including adenoviruses, poxviruses, rhabdoviruses, herpes viruses, paramyxovirus (PV) and reoviruses. Due to the difference in innate immune response to virus, anti-tumor mechanisms differ. Herpes viruses are DNA viruses capable of establishing lytic and latent infection in the host; the utility of herpes simplex virus 1 (HSV-1) as an oncolytic agent has been the most widely explored. Using gene editing method to make HSV-1 express GM-CSF can promote the recruitment of T cells to the tumor site and enhance the anti-tumor effect (147). PVs are members of the Paramyxoviridae family of disease-causing viruses in humans and animals. PV is a strong inducer of IFN and other immunostimulatory cytokines that activate various immune factors to mount excellent antitumor innate and adaptive immune responses (148). Mumps virus, which has been proven to have cytopathic effects, causes infected cells to secrete various cytokines and IFN pathway-associated genes or receptors to achieve anti-tumor effects (149).

The first FDA- and European Medicines Agency-approved oncolytic virus, talimogene laherparepvec (T-VEC), is a modified HSV virus for the treatment of malignant melanoma that encodes GM-CSF to enhance antitumor immune responses (150). A recombinant adenovirus (Oncorine®) was approved by Chinese regulators as early as November 2005 for the treatment of HNC (in combination with chemotherapy) and a number of oncolytic viruses are in clinical development (151,152). The multifunctional properties of oncolytic viruses in tumor therapy make them highly synergistic when used in combination with other drugs (153). Currently, a large body of evidence suggests that oncolytic virus therapy induces tumor cell death by enhancing the antigenicity of tumor cells or their susceptibility to immune cells when used in combination with radiotherapy, chemotherapy and other immunotherapies (154,155). Many naturally occurring viruses, such as parvovirus, measles virus, reovirus and Newcastle disease virus, exhibit a natural preference for cancer cells (156). However, other viruses such as adenovirus, vesicular stomatitis and vaccinia virus and HSV need to be engineered to be cancer specific (Table I). Four approaches are commonly used to design oncolytic viruses to selectively target tumor cells. The first is use of virus-specific receptor-mediated cellular targeting, such as EGFR and HER-2 (157). The second approach exploits the rapidly dividing nature of tumor cells to increase the efficiency of viral replication compared with normal cells. For example, mutations in tumor drivers or other enzymes such as protein kinase R increase viral replication in tumor cells (158). Numerous types of tumor cell exhibit a lack of normal antiviral IFN or TNF responses that promote selective viral replication (159). The fourth is that normal cells respond to viral infection by inducing apoptosis or inhibiting translation, transcription and/or transduction targeting to prevent cell lysis, which may limit viral spread (160).

Table I.

Drugs used in oncolytic virus anti-tumor clinical trials.

Table I.

Drugs used in oncolytic virus anti-tumor clinical trials.

Virus typeProductMethod of administrationCancer typeApproved
AdenovirusOnyx-015IntratumoralHead and neck, pancreatic, ovarian, colorectal, glioma, lung and liver metastasisYes
H101IntratumoralSquamous cell carcinoma, head and neck cancerYes
DNX-2401IntratumoralGlioblastoma, ovarianNo
VCN-01IntratumoralPancreaticNo
Colo-Ad1IntratumoralColon, non-small cell lung, renal, bladder, ovarianNo
ProstAtakIntratumoralPancreatic, lung, breast, mesothelioma, prostateNo
Oncos-102IntratumoralSolid tumorNo
CG0070IntratumoralBladderNo
ICOVIR5IntravenousMelanoma, solid tumorYes
Ad5-yCD/IntratumoralProstate, pancreaticNo
mutTKSR39-rephIL12
Ads/HSV-tkIntratumoralTriple-negative breast, non-small cell lungNo
LOAd703IntratumoralMalignant melanomaNo
TasadenoturevIntratumoralRecurrent gliomaNo
Vaccinia (JX-594)Pexa-vacIntratumoral, intravenousMelanoma, liver, colorectal, breast, hepatocellular carcinomaNo
GL-ONC1Intraperitoneal, intratumoral, intravenousLung, head and neck, mesotheliomaNo
HerpesT-VECIntratumoralMelanoma, head and neck, pancreaticYes
G207IntratumoralGlioblastomaYes
HF10IntratumoralBreast, melanoma, pancreaticYes
HSV1716IntratumoralHepatocellular carcinoma, glioblastoma, mesothelioma, neuroblastomaYes
OrienX010IntravenousGlioblastomaYes
ReovirusReolysinIntravenous, intratumoralGlioma, sarcoma, colorectal, non-small cell lung, ovarian, melanoma, pancreatic, multiple myeloma, head and neckYes
Seneca ValleySVV-001Intratumoral Neuroendocrine-featured tumor, neuroblastoma, lungYes
CoxsackievirusCavatakIntratumoralMelanoma, breast, prostateYes
(CVA21)
Newcastle diseasePV701IntravenousSquamous cell carcinoma of the larynx, salivary glandNo
NDV-HUJIntravenousGlioblastoma, sarcoma, neuroblastomaNo
Vesicular stomatitisVSV-hIFNβIntratumoralHead and neck squamous cell carcinoma, non-small cell lung, hepatocellular carcinomaNo
MeaslesMV-NISIntraperitoneal, intratumoralMyeloma, ovarian, mesothelioma, non-small cell lungNo
MV-CEAIntraperitoneal, IntratumoralGlioblastoma, ovarian, fallopian tube endometrioid adenocarcinomaNo

The immune response to oncolytic viruses is an important part of the antitumor effect, but it can be a double-edged sword. On the one hand, viruses promote immune responses against tumor cells by increasing tumor antigen presentation via viral infection. On the other hand, neutralizing antiviral responses may prevent viral replication and persistent infection of tumor cells (161). Therapeutic outcomes depend on the complex interplay between these opposing forces, and local injection of the tumor can be used to observe the therapeutic response. To balance this response of the immune system, methods to optimize current oncolytic viruses or develop novel viruses to enhance the stimulation of the host immune response to tumor cells without triggering rapid clearance of oncolytic viruses (for example, deletion of Herpes simplex virus protein ICP34.5 and ICP47 in T-VEC) have been investigated (162). In addition, genes for cytokines or chemokines can be integrated into the genome to enhance the therapeutic effect of oncolytic viruses. GM-CSF, an immune-associated cytokine, can increase APC activation and trigger systemic antitumor immune responses, which increases oncolytic virus efficacy (163). TAAs, immune-associated ligands, or bispecific T-cell engager antibodies can also be used to modify oncolytic viruses (164).

In addition, it is difficult for some viruses to obtain extremely high titer products required for clinical doses, which limits large-scale production of oncolytic viruses due to the high cost of current technologies (165). The host immune barrier and antiviral response can inhibit viral replication and lead to resistance to oncolytic viruses, such as macrophages, which directly capture viruses in organs such as the liver, thereby decreasing viral titers in the body and affecting oncolysis (166). The most common adverse reactions to oncolytic viruses are fever and local injection site reactions but can also include chills, nausea and vomiting, flu-like symptoms, fatigue and pain (167).

In conclusion, although oncolytic viruses have potential, there are some obstacles to their production and application. Current molecular biotechnology strategies enhance the targeting and killing effects of oncolytic viruses, but further research is needed to develop tumor treatments with higher efficacy and lower adverse reaction rates. The production technology of oncolytic virus is imperfect, and there is no uniform standard for industrial production quality and inspection, which is an obstacle to its application.

A recent strategy is to make tumors express ABO blood group antigens and using a mechanism similar to that caused by blood group incompatibility to activate the immune system to kill tumor cells (168). This differs from passive immunotherapy as it does not activate the immune response systemically; it also removes the need to identify tumor-associated antigens as it does not require the body to produce specific anti-tumor CTL cells. Different from the mechanism of oncolytic viruses, the virus does not directly lyse cells but serves as a carrier to express blood group antigens on the tumor cell membrane and is recognized and activated by naturally occurring blood group antibodies. The complement response produces cytolysis, which demonstrates the key anti-tumor role of the innate immune system (169). Although studies have detected loss of blood group antigen expression in primary breast tumors and their metastases, loss of blood group antigen expression may be considered a marker of invasion and half of proximal colon tumors show loss of antigen expression (170172). However, this does not simply activate the immune system by allowing the tumor to express blood group antigens. If a tumor expresses an antigen, there will be an antibody that can bind to it naturally in the body, and it will react with the antigen and antibody, and then activate the immune system to produce cell lysis. There are similar antigens that can treat tumors, such as the Rhesus blood group antigen. Expressing the corresponding antigens on tumor cells of patients with autoimmune diseases is also a strategy for treating tumors (173). The advantages of adopting such a strategy to treat tumors include lack of tumor resistance to treatment; the lentiviral vector itself has little immunogenicity and is not easily cleared by the body. Theoretically, as long as the tumor tissue expresses an antigen that can be recognized by the body's immune system, tumor cells can be directly recognized by the immune system and produce a lytic reaction. With the development of molecular biology, vector viruses can be used to make tumor cells express any protein. Therefore, this treatment method can solve the problem of tumor drug resistance. Intratumoral drug injection therapy is safer than systemic medication and can avoid the failure of body organs caused by the storm of inflammatory factors caused by systemic medication. Simple intratumoral injections decrease the risk and pain associated with surgery and chemotherapy; the procedure is simple and can be performed by doctors in the primary hospital. Although the local administration method is safer compared with systemic administration, the optimal injection dose still needs to be determined to ensure adequate dispersion of drug in the tumor tissue and minimize the leakage from the tumor tissue. As a novel tumor treatment strategy, further research is required to develop use of naturally occurring antigen-antibody immune response to treat tumors.

Conclusion

In the past decades, anti-cancer immunotherapy has changed from a promising treatment method to a reality of clinical treatment. Many immunotherapy programs that can be used for patients with cancer have now been approved by the US FDA and European Medicines Agency and many other treatment programs are being studied as independent therapeutic interventions or in combination with clinical routine treatment (174,175). Treatment strategies are no longer based solely on interfering with metabolism of tumor cells or whether it is a purely clonal proliferative disease. A number of studies has shown that the survival of tumor cells depends to a large extent on the surrounding environment, which contains abundant and heterogeneous untransformed components, including stroma and endothelial and immune cells (176178).

Immunotherapy has become a clinical reality, and an increasing number of patients with cancer will receive immunotherapy at some stage. The treatment of tumors by interfering with immune checkpoints has become an important and effective form of immunotherapy. Drugs targeting CTLA-4, PD-1 and PD-L1 are the most widely studied (179). Numerous studies have also shown that what was previously classified as passive immunotherapy, including several tumor-targeting monoclonal antibodies, adoptive T cell transfer, and oncolytic viruses, may constitute a potent active form of immunotherapy (115,180,181). Drugs such as immunosuppressive metabolic inhibitors and PRR agonists have attracted interest not only as adjuvants to conventional vaccines, but also as therapeutic measures that may mediate the antitumor effect or enhance the therapeutic effects of other anticancer drugs (182).

In 2013, the clinical success of immunotherapy was named ‘Breakthrough of the Year’ by Science (183). Clinical research is also focused on whether immunotherapy can be used as a stand-alone treatment or in combination with other antitumor drugs to improve the efficacy and safety in patients with cancer.

One of the key challenges in developing cancer vaccines is to identify specific tumor antigens for use as immunotherapeutic targets. Good target antigens should exhibit high antigenicity and homologous expression in tumor tissue to overcome problems caused by tumor heterogeneity. Tumor cells can undergo antigen modulation, which means that the immune system attacks tumor cells, resulting in reduction or loss of tumor antigen epitopes on the surface, thereby escaping recognition and killing by the immune system (32,184). Studies have also confirmed that tumor cells exhibit characteristics of stem cells and may actively decrease expression of antigens (185187). The development of therapeutic oncolytic viruses is faced with challenges regarding how to formulate a reasonable clinical trial design, dosing regimen, pharmacodynamic analysis and biosafety issues (188). Immunocompromised patients may not be candidates for oncolytic virus therapy because oncolytic virus-mediated antitumor immunity may be compromised in these patients (189). The treatment of tumors with blood group antigens is different from that of oncolytic viruses; it actively allows tumors to express naturally occurring antigens in the body to activate the natural immune response and treat tumors (190). Further studies are required to determine clinical feasibility.

In summary, tumor immunotherapy may enhance the direct killing effect of CTLs on tumors and weaken the immunosuppressive TME.

Acknowledgements

Not applicable.

Funding

The present study was supported by Inner Mongolia Natural Science Foundation Project (grant no. 2021MS08060) and Inner Mongolia Medical University Joint Project (grant no. YKD2021LH068).

Availability of data and materials

Not applicable.

Authors' contributions

FZ, DS and LW performed the literature review and wrote the manuscript. MS, HY, XZ, LC and ZH wrote the manuscript. LL and LW conceived, reviewed and revised the article. All authors revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

LW is the corresponding author of ABO blood group antigen therapy: A potential new strategy against solid tumors. The other authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

cDC

conventional dendritic cell

CTLA-4

cytotoxic T lymphocyte-associated antigen 4

FDA

Food and Drug Administration

NK

natural killer

HSV

herpes simplex virus

PD-1

programmed death receptor 1

CAR

chimeric antigen receptor

TCR

T cell receptor

APC

antigen-presenting cell

TME

tumor microenvironment

mAb

monoclonal antibody

GM-CSF

granulocyte macrophage colony-stimulating factor

References

1 

Lin L, Li Z, Yan L, Liu Y, Yang H and Li H: Global, regional, and national cancer incidence and death for 29 cancer groups in 2019 and trends analysis of the global cancer burden, 1990–2019. J Hematol Oncol. 14:1972021. View Article : Google Scholar : PubMed/NCBI

2 

Feng RM, Zong YN, Cao SM and Xu RH: Current cancer situation in China: Good or bad news from the 2018 global cancer statistics? Cancer Commun (Lond). 39:222019. View Article : Google Scholar : PubMed/NCBI

3 

Pan C, Liu H, Robins E, Song W, Liu D, Li Z and Zheng L: Next-generation immuno-oncology agents: Current momentum shifts in cancer immunotherapy. J Hematol Oncol. 13:292020. View Article : Google Scholar : PubMed/NCBI

4 

Sun Y, Jiang L, Wen T, Guo X, Shao X, Qu H, Chen X, Song Y, Wang F, Qu X and Li Z: Trends in the research into immune checkpoint blockade by anti-PD1/PDL1 antibodies in cancer immunotherapy: A bibliometric study. Front Pharmacol. 12:6709002021. View Article : Google Scholar : PubMed/NCBI

5 

Zhang Y, Du X, Liu M, Tang F, Zhang P, Ai C, Fields JK, Sundberg EJ, Latinovic OS, Devenport M, et al: Hijacking antibody-induced CTLA-4 lysosomal degradation for safer and more effective cancer immunotherapy. Cell Res. 9:609–627. 2019. View Article : Google Scholar

6 

Smith KM and Desai J: Nivolumab for the treatment of colorectal cancer. Expert Rev Anticancer Ther. 18:611–618. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Gonzalez H, Hagerling C and Werb Z: Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 32:1267–1284. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Ribatti D: The concept of immune surveillance against tumors. The first theories. Oncotarget. 8:7175–7180. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Dunn GP, Bruce AT, Ikeda H, Old LJ and Schreiber RD: Cancer immunoediting: From immunosurveillance to tumor escape. Nat Immunol. 3:991–998. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Mittal D, Gubin MM, Schreiber RD and Smyth MJ: New insights into cancer immunoediting and its three component phases-elimination, equilibrium and escape. Curr Opin Immunol. 27:16–25. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Finn OJ: Human tumor antigens yesterday, today, and tomorrow. Cancer Immunol Res. 5:347–354. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Verginelli F, Pisacane A, Gambardella G, D'Ambrosio A, Candiello E, Ferrio M, Panero M, Casorzo L, Benvenuti S, Cascardi E, et al: Cancer of unknown primary stem-like cells model multi-organ metastasis and unveil liability to MEK inhibition. Nat Commun. 12:24982021. View Article : Google Scholar : PubMed/NCBI

13 

Deng H, Yang W, Zhou Z, Tian R, Lin L, Ma Y, Song J and Chen X: Targeted scavenging of extracellular ROS relieves suppressive immunogenic cell death. Nat Commun. 11:49512020. View Article : Google Scholar : PubMed/NCBI

14 

Ren Y, Kumar A, Das JK, Peng HY, Wang L, Balllard D, Xiong X, Ren X, Zhang Y, Yang JM and Song J: Tumorous expression of NAC1 restrains antitumor immunity through the LDHA-mediated immune evasion. J Immunother Cancer. 10:e0048562022. View Article : Google Scholar : PubMed/NCBI

15 

Bazett M, Costa AM, Bosiljcic M, Anderson RM, Alexander MP, Wong SWY, Dhanji S, Chen JM, Pankovich J, Lam S, et al: Harnessing innate lung anti-cancer effector functions with a novel bacterial-derived immunotherapy. Oncoimmunology. 7:e13988752017. View Article : Google Scholar : PubMed/NCBI

16 

Rosewell Shaw A, Porter C, Biegert G, Jatta L and Suzuki M: HydrAd: A helper-dependent adenovirus targeting multiple immune pathways for cancer immunotherapy. Cancers (Basel). 14:27692022. View Article : Google Scholar : PubMed/NCBI

17 

Zhang L, Li Z, Skrzypczynska KM, Fang Q, Zhang W, O'Brien SA, He Y, Wang L, Zhang Q, Kim A, et al: Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell. 181:442–459.e29. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Krausgruber T, Fortelny N, Fife-Gernedl V, Senekowitsch M, Schuster LC, Lercher A, Nemc A, Schmidl C, Rendeiro AF, Bergthaler A and Bock C: Structural cells are key regulators of organ-specific immune responses. Nature. 583:296–302. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Ahmad HI, Jabbar A, Mushtaq N, Javed Z, Hayyat MU, Bashir J, Naseeb I, Abideen ZU, Ahmad N and Chen J: Immune tolerance vs immune resistance: The interaction between host and pathogens in infectious diseases. Front Vet Sci. 9:8274072022. View Article : Google Scholar : PubMed/NCBI

20 

Bettcher BM, Tansey MG, Dorothée G and Heneka MT: Peripheral and central immune system crosstalk in Alzheimer disease-a research prospectus. Nat Rev Neurol. 17:689–701. 2021. View Article : Google Scholar : PubMed/NCBI

21 

Morsink MAJ, Willemen NGA, Leijten J, Bansal R and Shin SR: Immune organs and immune cells on a chip: An overview of biomedical applications. Micromachines (Basel). 11:8492020. View Article : Google Scholar : PubMed/NCBI

22 

Benci JL, Johnson LR, Choa R, Xu Y, Qiu J, Zhou Z, Xu B, Ye D, Nathanson KL, June CH, et al: Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell. 178:933–948.e14. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Gangopadhyay K, Roy S, Sen Gupta S, Chandradasan AC, Chowdhury S and Das R: Regulating the discriminatory response to antigen by T-cell receptor. Biosci Rep. 42:BSR202120122022. View Article : Google Scholar : PubMed/NCBI

24 

Druszczyńska M, Godkowicz M, Kulesza J, Wawrocki S and Fol M: Cytokine receptors-regulators of antimycobacterial immune response. Int J Mol Sci. 23:11122022. View Article : Google Scholar : PubMed/NCBI

25 

Šket T, Ramuta TŽ, Starčič Erjavec M and Kreft ME: The role of innate immune system in the human amniotic membrane and human amniotic fluid in protection against intra-amniotic infections and inflammation. Front Immunol. 12:7353242021. View Article : Google Scholar : PubMed/NCBI

26 

He L, Valignat MP, Zhang L, Gelard L, Zhang F, Le Guen V, Audebert S, Camoin L, Fossum E, Bogen B, et al: ARHGAP45 controls naïve T- and B-cell entry into lymph nodes and T-cell progenitor thymus seeding. EMBO Rep. 22:e521962021. View Article : Google Scholar : PubMed/NCBI

27 

Yiwen Z, Shilin G, Yingshi C, Lishi S, Baohong L, Chao L, Linghua L, Ting P and Hui Z: Efficient generation of antigen-specific CTLs by the BAFF-activated human B Lymphocytes as APCs: A novel approach for immunotherapy. Oncotarget. 7:77732–77748. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Nicholson LB: The immune system. Essays Biochem. 60:275–301. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Vila-Leahey A, MacKay A, Portales-Cervantes L, Weir GM, Merkx-Jacques A and Stanford MM: Generation of highly activated, antigen-specific tumor-infiltrating CD8+ T cells induced by a novel T cell-targeted immunotherapy. Oncoimmunology. 9:17825742020. View Article : Google Scholar : PubMed/NCBI

30 

Schönefeldt S, Wais T, Herling M, Mustjoki S, Bekiaris V, Moriggl R and Neubauer HA: The diverse roles of γδ T cells in cancer: From rapid immunity to aggressive lymphoma. Cancers (Basel). 13:62122021. View Article : Google Scholar : PubMed/NCBI

31 

Leko V and Rosenberg SA: Identifying and targeting human tumor antigens for T cell-based immunotherapy of solid tumors. Cancer Cell. 38:454–472. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Liu J, Fu M, Wang M, Wan D, Wei Y and Wei X: Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. J Hematol Oncol. 15:282022. View Article : Google Scholar : PubMed/NCBI

33 

Liu YT and Sun ZJ: Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 11:5365–5386. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Tanimine N, Ohira M, Tahara H, Ide K, Tanaka Y, Onoe T and Ohdan H: Strategies for deliberate induction of immune tolerance in liver transplantation: From preclinical models to clinical application. Front Immunol. 11:16152020. View Article : Google Scholar : PubMed/NCBI

35 

Hargadon KM: Tumor microenvironmental influences on dendritic cell and T cell function: A focus on clinically relevant immunologic and metabolic checkpoints. Clin Transl Med. 10:374–411. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Elizondo DM, Andargie TE, Kubhar DS, Gugssa A and Lipscomb MW: CD40-CD40L cross-talk drives fascin expression in dendritic cells for efficient antigen presentation to CD4+ T cells. Int Immunol. 29:121–131. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Morris DL, Oatmen KE, Mergian TA, Cho KW, DelProposto JL, Singer K, Evans-Molina C, O'Rourke RW and Lumeng CN: CD40 promotes MHC class II expression on adipose tissue macrophages and regulates adipose tissue CD4+ T cells with obesity. J Leukoc Biol. 99:1107–1119. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Zhou Y, Slone N, Chrisikos TT, Kyrysyuk O, Babcock RL, Medik YB, Li HS, Kleinerman ES and Watowich SS: Vaccine efficacy against primary and metastatic cancer with in vitro-generated CD103+ conventional dendritic cells. J Immunother Cancer. 8:e0004742020. View Article : Google Scholar : PubMed/NCBI

39 

Krishnaswamy JK, Gowthaman U, Zhang B, Mattsson J, Szeponik L, Liu D, Wu R, White T, Calabro S, Xu L, et al: Migratory CD11b+ conventional dendritic cells induce T follicular helper cell-dependent antibody responses. Sci Immunol. 2:eaam91692017. View Article : Google Scholar : PubMed/NCBI

40 

Roselli E, Araya P, Núñez NG, Gatti G, Graziano F, Sedlik C, Benaroch P, Piaggio E and Maccioni M: TLR3 activation of intratumoral CD103+ dendritic cells modifies the tumor infiltrate conferring anti-tumor immunity. Front Immunol. 10:5032019. View Article : Google Scholar : PubMed/NCBI

41 

Koh YC, Yang G, Lai CS, Weerawatanakorn M and Pan MH: Chemopreventive effects of phytochemicals and medicines on M1/M2 polarized macrophage role in inflammation-related diseases. Int J Mol Sci. 19:22082018. View Article : Google Scholar : PubMed/NCBI

42 

Castro F, Cardoso AP, Gonçalves RM, Serre K and Oliveira MJ: Interferon-gamma at the crossroads of tumor immune surveillance or evasion. Front Immunol. 9:8472018. View Article : Google Scholar : PubMed/NCBI

43 

Song X, Traub B, Shi J and Kornmann M: Possible roles of interleukin-4 and −13 and their receptors in gastric and colon cancer. Int J Mol Sci. 22:7272021. View Article : Google Scholar : PubMed/NCBI

44 

Babazadeh S, Nassiri SM, Siavashi V, Sahlabadi M, Hajinasrollah M and Zamani-Ahmadmahmudi M: Macrophage polarization by MSC-derived CXCL12 determines tumor growth. Cell Mol Biol Lett. 26:302021. View Article : Google Scholar : PubMed/NCBI

45 

Oshi M, Tokumaru Y, Asaoka M, Yan L, Satyananda V, Matsuyama R, Matsuhashi N, Futamura M, Ishikawa T, Yoshida K, et al: M1 Macrophage and M1/M2 ratio defined by transcriptomic signatures resemble only part of their conventional clinical characteristics in breast cancer. Sci Rep. 10:165542020. View Article : Google Scholar : PubMed/NCBI

46 

He M, Wang Y, Zhang G, Cao K, Yang M and Liu H: The prognostic significance of tumor-infiltrating lymphocytes in cervical cancer. J Gynecol Oncol. 32:e322021. View Article : Google Scholar : PubMed/NCBI

47 

Xu A, Zhang L, Yuan J, Babikr F, Freywald A, Chibbar R, Moser M, Zhang W, Zhang B, Fu Z and Xiang J: TLR9 agonist enhances radiofrequency ablation-induced CTL responses, leading to the potent inhibition of primary tumor growth and lung metastasis. Cell Mol Immunol. 16:820–832. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Crawshaw A, Kendrick YR, McMichael AJ and Ho LP: Abnormalities in iNKT cells are associated with impaired ability of monocytes to produce IL-10 and suppress T-cell proliferation in sarcoidosis. Eur J Immunol. 44:2165–2174. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Wang YL, Gong Y, Lv Z, Li L and Yuan Y: Expression of PD1/PDL1 in gastric cancer at different microsatellite status and its correlation with infiltrating immune cells in the tumor microenvironment. J Cancer. 12:1698–1707. 2021. View Article : Google Scholar : PubMed/NCBI

50 

Weigelin B, Bolaños E, Teijeira A, Martinez-Forero I, Labiano S, Azpilikueta A, Morales-Kastresana A, Quetglas JI, Wagena E, Sánchez-Paulete AR, et al: Focusing and sustaining the antitumor CTL effector killer response by agonist anti-CD137 mAb. Proc Natl Acad Sci USA. 112:7551–7556. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Baik AH, Tsai KK, Oh DY and Aras MA: Mechanisms and clinical manifestations of cardiovascular toxicities associated with immune checkpoint inhibitors. Clin Sci (Lond). 135:703–724. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Turula H, Bragazzi Cunha J, Mainou BA, Ramakrishnan SK, Wilke CA, Gonzalez-Hernandez MB, Pry A, Fava J, Bassis CM, Edelman J, et al: Natural secretory immunoglobulins promote enteric viral infections. J Virol. 92:e00826–18. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Ehrlich P: Address in pathology, on chemiotherapy: Delivered before the seventeenth international congress of medicine. Br Med J. 2:353–359. 1913. View Article : Google Scholar : PubMed/NCBI

54 

He Q, Liu Z, Liu Z, Lai Y, Zhou X and Weng J: TCR-like antibodies in cancer immunotherapy. J Hematol Oncol. 12:992019. View Article : Google Scholar : PubMed/NCBI

55 

Byeon HK, Ku M and Yang J: Beyond EGFR inhibition: Multilateral combat strategies to stop the progression of head and neck cancer. Exp Mol Med. 51:1–14. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Santoro V, Jia R, Thompson H, Nijhuis A, Jeffery R, Kiakos K, Silver AR, Hartley JA and Hochhauser D: Role of reactive oxygen species in the abrogation of oxaliplatin activity by cetuximab in colorectal cancer. J Natl Cancer Inst. 108:djv3942015. View Article : Google Scholar : PubMed/NCBI

57 

Chen KF, Chen HL, Shiau CW, Liu CY, Chu PY, Tai WT, Ichikawa K, Chen PJ and Cheng AL: Sorafenib and its derivative SC-49 sensitize hepatocellular carcinoma cells to CS-1008, a humanized anti-TNFRSF10B (DR5) antibody. Br J Pharmacol. 168:658–672. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Fenwarth L, Fournier E, Cheok M, Boyer T, Gonzales F, Castaigne S, Boissel N, Lambert J, Dombret H, Preudhomme C and Duployez N: Biomarkers of gemtuzumab ozogamicin response for acute myeloid leukemia treatment. Int J Mol Sci. 21:56262020. View Article : Google Scholar : PubMed/NCBI

59 

Boyer-Suavet S, Andreani M, Lateb M, Savenkoff B, Brglez V, Benzaken S, Bernard G, Nachman PH, Esnault V and Seitz-Polski B: Neutralizing anti-rituximab antibodies and relapse in membranous nephropathy treated with rituximab. Front Immunol. 10:30692020. View Article : Google Scholar : PubMed/NCBI

60 

Strohl WR and Naso M: Bispecific T-Cell redirection versus chimeric antigen receptor (CAR)-T cells as approaches to kill cancer cells. Antibodies (Basel). 8:412019. View Article : Google Scholar : PubMed/NCBI

61 

Huo Y, Sheng Z, Lu DR, Ellwanger DC, Li CM, Homann O, Wang S, Yin H and Ren R: Blinatumomab-induced T cell activation at single cell transcriptome resolution. BMC Genomics. 22:1452021. View Article : Google Scholar : PubMed/NCBI

62 

Davis SK, Selva KJ, Kent SJ and Chung AW: Serum IgA Fc effector functions in infectious disease and cancer. Immunol Cell Biol. 98:276–286. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Brandsma AM, Bondza S, Evers M, Koutstaal R, Nederend M, Jansen JHM, Rösner T, Valerius T, Leusen JHW and Ten Broeke T: Potent Fc receptor signaling by IgA leads to superior killing of cancer cells by neutrophils compared to IgG. Front Immunol. 10:7042019. View Article : Google Scholar : PubMed/NCBI

64 

Leoh LS, Daniels-Wells TR and Penichet ML: IgE immunotherapy against cancer. Curr Top Microbiol Immunol. 388:109–149. 2015.PubMed/NCBI

65 

Chauhan J, McCraw AJ, Nakamura M, Osborn G, Sow HS, Cox VF, Stavraka C, Josephs DH, Spicer JF, Karagiannis SN and Bax HJ: IgE antibodies against cancer: Efficacy and safety. Antibodies (Basel). 9:552020. View Article : Google Scholar : PubMed/NCBI

66 

El-Kadiry AE, Rafei M and Shammaa R: Cell therapy: Types, regulation, and clinical benefits. Front Med (Lausanne). 8:7560292021. View Article : Google Scholar : PubMed/NCBI

67 

Mora Román JJ, Del Campo M, Villar J, Paolini F, Curzio G, Venuti A, Jara L, Ferreira J, Murgas P, Lladser A, et al: Immunotherapeutic potential of mollusk hemocyanins in combination with human vaccine adjuvants in murine models of oral cancer. J Immunol Res. 2019:70769422019. View Article : Google Scholar : PubMed/NCBI

68 

Kurosawa S, Mizuno S, Arai Y, Masuko M, Kanda J, Kohno K, Onai D, Fukuda T, Ozawa Y, Katayama Y, et al: Syngeneic hematopoietic stem cell transplantation for acute myeloid leukemia: A propensity score-matched analysis. Blood Cancer J. 11:1592021. View Article : Google Scholar : PubMed/NCBI

69 

Lin J, Wang H, Liu C, Cheng A, Deng Q, Zhu H and Chen J: Dendritic cells: Versatile players in renal transplantation. Front Immunol. 12:6545402021. View Article : Google Scholar : PubMed/NCBI

70 

Markov O, Oshchepkova A and Mironova N: Immunotherapy based on dendritic cell-targeted/-derived extracellular vesicles-A novel strategy for enhancement of the anti-tumor immune response. Front Pharmacol. 10:11522019. View Article : Google Scholar : PubMed/NCBI

71 

Blando J, Sharma A, Higa MG, Zhao H, Vence L, Yadav SS, Kim J, Sepulveda AM, Sharp M, Maitra A, et al: Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer. Proc Natl Acad Sci USA. 116:1692–1697. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Cui X and Snapper CM: Epstein barr virus: Development of vaccines and immune cell therapy for EBV-associated diseases. Front Immunol. 12:7344712021. View Article : Google Scholar : PubMed/NCBI

73 

Luo XH, Meng Q, Rao M, Liu Z, Paraschoudi G, Dodoo E and Maeurer M: The impact of inflationary cytomegalovirus-specific memory T cells on anti-tumour immune responses in patients with cancer. Immunology. 155:294–308. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Pesch T, Bonati L, Kelton W, Parola C, Ehling RA, Csepregi L, Kitamura D and Reddy ST: Molecular design, optimization, and genomic integration of chimeric B cell receptors in murine B cells. Front Immunol. 10:26302019. View Article : Google Scholar : PubMed/NCBI

75 

Stoiber S, Cadilha BL, Benmebarek MR, Lesch S, Endres S and Kobold S: Limitations in the design of chimeric antigen receptors for cancer therapy. Cells. 8:4722019. View Article : Google Scholar : PubMed/NCBI

76 

Kasala D, Yoon AR, Hong J, Kim SW and Yun CO: Evolving lessons on nanomaterial-coated viral vectors for local and systemic gene therapy. Nanomedicine (Lond). 11:1689–1713. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Muliaditan T, Halim L, Whilding LM, Draper B, Achkova DY, Kausar F, Glover M, Bechman N, Arulappu A, Sanchez J, et al: Synergistic T cell signaling by 41BB and CD28 is optimally achieved by membrane proximal positioning within parallel chimeric antigen receptors. Cell Rep Med. 2:1004572021. View Article : Google Scholar : PubMed/NCBI

78 

Verheye E, Bravo Melgar J, Deschoemaeker S, Raes G, Maes A, De Bruyne E, Menu E, Vanderkerken K, Laoui D and De Veirman K: Dendritic cell-based immunotherapy in multiple myeloma: Challenges, opportunities, and future directions. Int J Mol Sci. 23:9042022. View Article : Google Scholar : PubMed/NCBI

79 

Steinman RM and Cohn ZA: Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med. 137:1142–1162. 1973. View Article : Google Scholar : PubMed/NCBI

80 

Steinman RM and Banchereau J: Taking dendritic cells into medicine. Nature. 449:419–426. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Handy CE and Antonarakis ES: Sipuleucel-T for the treatment of prostate cancer: Novel insights and future directions. Future Oncol. 14:907–917. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Artyomov MN, Munk A, Gorvel L, Korenfeld D, Cella M, Tung T and Klechevsky E: Modular expression analysis reveals functional conservation between human Langerhans cells and mouse cross-priming dendritic cells. J Exp Med. 212:743–757. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Barbosa CRR, Barton J, Shepherd AJ and Mishto M: Mechanistic diversity in MHC class I antigen recognition. Biochem J. 478:4187–4202. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Oh DS and Lee HK: Autophagy protein ATG5 regulates CD36 expression and anti-tumor MHC class II antigen presentation in dendritic cells. Autophagy. 15:2091–2106. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Rühle PF, Fietkau R, Gaipl US and Frey B: Development of a modular assay for detailed immunophenotyping of peripheral human whole blood samples by multicolor flow cytometry. Int J Mol Sci. 17:13162016. View Article : Google Scholar : PubMed/NCBI

86 

Mengos AE, Gastineau DA and Gustafson MP: The CD14+HLA-DRlo/neg monocyte: An immunosuppressive phenotype that restrains responses to cancer immunotherapy. Front Immunol. 10:11472019. View Article : Google Scholar : PubMed/NCBI

87 

Trujillo-Ocampo A, Cho HW, Clowers M, Pareek S, Ruiz-Vazquez W, Lee SE and Im JS: IL-7 during antigenic stimulation using allogeneic dendritic cells promotes expansion of CD45RACD62L+CD4+ invariant NKT cells with Th-2 biased cytokine production profile. Front Immunol. 11:5674062020. View Article : Google Scholar : PubMed/NCBI

88 

Mylonas KJ, Anderson J, Sheldrake TA, Hesketh EE, Richards JA, Ferenbach DA, Kluth DC, Savill J and Hughes J: Granulocyte macrophage-colony stimulating factor: A key modulator of renal mononuclear phagocyte plasticity. Immunobiology. 224:60–74. 2019. View Article : Google Scholar : PubMed/NCBI

89 

Shklovskaya E and Rizos H: MHC class I deficiency in solid tumors and therapeutic strategies to overcome it. Int J Mol Sci. 22:67412021. View Article : Google Scholar : PubMed/NCBI

90 

Pardi N, Hogan MJ, Porter FW and Weissman D: mRNA vaccines-a new era in vaccinology. Nat Rev Drug Discov. 17:261–279. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Coughlan L, Kremer EJ and Shayakhmetov DM: Adenovirus-based vaccines-a platform for pandemic preparedness against emerging viral pathogens. Mol Ther. 30:1822–1849. 2022. View Article : Google Scholar : PubMed/NCBI

92 

Guo J, De May H, Franco S, Noureddine A, Tang L, Brinker CJ, Kusewitt DF, Adams SF and Serda RE: Cancer vaccines from cryogenically silicified tumour cells functionalized with pathogen-associated molecular patterns. Nat Biomed Eng. 6:19–31. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Subtil B, Cambi A, Tauriello DVF and de Vries IJM: The therapeutic potential of tackling tumor-induced dendritic cell dysfunction in colorectal cancer. Front Immunol. 12:7248832021. View Article : Google Scholar : PubMed/NCBI

94 

Guo L, Sang M, Liu Q, Fan X, Zhang X and Shan B: The expression and clinical significance of melanoma-associated antigen-A1, -A3 and -A11 in glioma. Oncol Lett. 6:55–62. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Gopanenko AV, Kosobokova EN and Kosorukov VS: Main strategies for the identification of neoantigens. Cancers (Basel). 12:28792020. View Article : Google Scholar : PubMed/NCBI

96 

Tomić S, Petrović A, Puač N, Škoro N, Bekić M, Petrović ZL and Čolić M: Plasma-activated medium potentiates the immunogenicity of tumor cell lysates for dendritic cell-based cancer vaccines. Cancers (Basel). 13:16262021. View Article : Google Scholar : PubMed/NCBI

97 

Polyzoidis S and Ashkan K: DCVax®-L-developed by northwest biotherapeutics. Hum Vaccin Immunother. 10:3139–3145. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Mott KR, Allen SJ, Zandian M, Akbari O, Hamrah P, Maazi H, Wechsler SL, Sharpe AH, Freeman GJ and Ghiasi H: Inclusion of CD80 in HSV targets the recombinant virus to PD-L1 on DCs and allows productive infection and robust immune responses. PLoS One. 9:e876172014. View Article : Google Scholar : PubMed/NCBI

99 

Stewart MP, Langer R and Jensen KF: Intracellular delivery by membrane disruption: Mechanisms, strategies, and concepts. Chem Rev. 118:7409–7531. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Bello MB, Yusoff K, Ideris A, Hair-Bejo M, Jibril AH, Peeters BPH and Omar AR: Exploring the prospects of engineered Newcastle disease virus in modern vaccinology. Viruses. 12:4512020. View Article : Google Scholar : PubMed/NCBI

101 

Leroy H, Han M, Woottum M, Bracq L, Bouchet J, Xie M and Benichou S: Virus-mediated cell-cell fusion. Int J Mol Sci. 21:96442020. View Article : Google Scholar : PubMed/NCBI

102 

Russell CJ and Hurwitz JL: Sendai virus-vectored vaccines that express envelope glycoproteins of respiratory viruses. Viruses. 13:10232021. View Article : Google Scholar : PubMed/NCBI

103 

Huang TX, Tan XY, Huang HS, Li YT, Liu BL, Liu KS, Chen X, Chen Z, Guan XY, Zou C and Fu L: Targeting cancer-associated fibroblast-secreted WNT2 restores dendritic cell-mediated antitumour immunity. Gut. 71:333–344. 2022. View Article : Google Scholar : PubMed/NCBI

104 

Cao L, Li W, Yang X, Zhang W, Li M, Zhang H, Qin C, Chen X and Gao R: Inhibition of host Ogr1 enhances effector CD8+ T-cell function by modulating acidic microenvironment. Cancer Gene Ther. 28:1213–1224. 2021. View Article : Google Scholar : PubMed/NCBI

105 

Aguilar-Cazares D, Chavez-Dominguez R, Marroquin-Muciño M, Perez-Medina M, Benito-Lopez JJ, Camarena A, Rumbo-Nava U and Lopez-Gonzalez JS: The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment. Front Endocrinol (Lausanne). 13:9295722022. View Article : Google Scholar : PubMed/NCBI

106 

Mizui M: Natural and modified IL-2 for the treatment of cancer and autoimmune diseases. Clin Immunol. 206:63–70. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Glassman CR, Mathiharan YK, Jude KM, Su L, Panova O, Lupardus PJ, Spangler JB, Ely LK, Thomas C, Skiniotis G and Garcia KC: Structural basis for IL-12 and IL-23 receptor sharing reveals a gateway for shaping actions on T versus NK cells. Cell. 184:983–999.e24. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Takeda K, Nakayama M, Hayakawa Y, Kojima Y, Ikeda H, Imai N, Ogasawara K, Okumura K, Thomas DM and Smyth MJ: IFN-γ is required for cytotoxic T cell-dependent cancer genome immunoediting. Nat Commun. 8:146072017. View Article : Google Scholar : PubMed/NCBI

109 

Duncan TJ, Rolland P, Deen S, Scott IV, Liu DT, Spendlove I and Durrant LG: Loss of IFN gamma receptor is an independent prognostic factor in ovarian cancer. Clin Cancer Res. 13:4139–4145. 2007. View Article : Google Scholar : PubMed/NCBI

110 

Ravaud A, Négrier S, Cany L, Merrouche Y, Le Guillou M, Blay JY, Clavel M, Gaston R, Oskam R and Philip T: Subcutaneous low-dose recombinant interleukin 2 and alpha-interferon in patients with metastatic renal cell carcinoma. Br J Cancer. 69:1111–1114. 1994. View Article : Google Scholar : PubMed/NCBI

111 

Weinreich DM and Rosenberg SA: Response rates of patients with metastatic melanoma to high-dose intravenous interleukin-2 after prior exposure to alpha-interferon or low-dose interleukin-2. J Immunother. 25:185–187. 2002. View Article : Google Scholar : PubMed/NCBI

112 

Wu YN, Zhang L, Chen T, Li X, He LH and Liu GX: Granulocyte-macrophage colony-stimulating factor protects mice against hepatocellular carcinoma by ameliorating intestinal dysbiosis and attenuating inflammation. World J Gastroenterol. 26:5420–5436. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Goyal G, Wong K, Nirschl CJ, Souders N, Neuberg D, Anandasabapathy N and Dranoff G: PPARγ contributes to immunity induced by cancer cell vaccines that secrete GM-CSF. Cancer Immunol Res. 6:723–732. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Zhang SR, Nold MF, Tang SC, Bui CB, Nold CA, Arumugam TV, Drummond GR, Sobey CG and Kim HA: IL-37 increases in patients after ischemic stroke and protects from inflammatory brain injury, motor impairment and lung infection in mice. Sci Rep. 9:69222019. View Article : Google Scholar : PubMed/NCBI

115 

Jones DS II, Nardozzi JD, Sackton KL, Ahmad G, Christensen E, Ringgaard L, Chang DK, Jaehger DE, Konakondla JV, Wiinberg M, et al: Cell surface-tethered IL-12 repolarizes the tumor immune microenvironment to enhance the efficacy of adoptive T cell therapy. Sci Adv. 8:eabi80752022. View Article : Google Scholar : PubMed/NCBI

116 

Miyazaki T, Maiti M, Hennessy M, Chang T, Kuo P, Addepalli M, Obalapur P, Sheibani S, Wilczek J, Pena R, et al: NKTR-255, a novel polymer-conjugated rhIL-15 with potent antitumor efficacy. J Immunother Cancer. 9:e0020242021. View Article : Google Scholar : PubMed/NCBI

117 

Hamilton JA: GM-CSF in inflammation. J Exp Med. 217:e201909452020. View Article : Google Scholar : PubMed/NCBI

118 

Kumar V: Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int Immunopharmacol. 89:1070872020. View Article : Google Scholar : PubMed/NCBI

119 

Mercogliano MF, Bruni S, Mauro F, Elizalde PV and Schillaci R: Harnessing Tumor necrosis factor alpha to achieve effective cancer immunotherapy. Cancers (Basel). 13:5642021. View Article : Google Scholar : PubMed/NCBI

120 

Wang TT, Zhao YL, Peng LS, Chen N, Chen W, Lv YP, Mao FY, Zhang JY, Cheng P, Teng YS, et al: Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut. 66:1900–1911. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Bi E, Ma X, Lu Y, Yang M, Wang Q, Xue G, Qian J, Wang S and Yi Q: Foxo1 and Foxp1 play opposing roles in regulating the differentiation and antitumor activity of TH9 cells programmed by IL-7. Sci Signal. 10:eaak97412017. View Article : Google Scholar : PubMed/NCBI

122 

Alizadeh D, Wong RA, Yang X, Wang D, Pecoraro JR, Kuo CF, Aguilar B, Qi Y, Ann DK, Starr R, et al: IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol Res. 7:759–772. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Hu HJ, Liang X, Li HL, Du CM, Hao JL, Wang HY, Gu JF, Ni AM, Sun LY, Xiao J, et al: The armed oncolytic adenovirus ZD55-IL-24 eradicates melanoma by turning the tumor cells from the self-state into the nonself-state besides direct killing. Cell Death Dis. 11:10222020. View Article : Google Scholar : PubMed/NCBI

124 

Johnson DE, O'Keefe RA and Grandis JR: Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 15:234–248. 2018. View Article : Google Scholar : PubMed/NCBI

125 

Park A, Kim S, Jung IH and Byun JH: An immune therapy model for effective treatment on inflammatory bowel disease. PLoS One. 15:e02389182020. View Article : Google Scholar : PubMed/NCBI

126 

Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, Pérez-Gracia JL, Rodríguez-Ruiz ME, Ponz-Sarvise M, Castañón E and Melero I: Cytokines in clinical cancer immunotherapy. Br J Cancer. 120:6–15. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Waldmann TA: Cytokines in cancer immunotherapy. Cold Spring Harb Perspect Biol. 10:a0284722018. View Article : Google Scholar : PubMed/NCBI

128 

Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA and Kobold S: Interleukins in cancer: From biology to therapy. Nat Rev Cancer. 21:481–499. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Riley P, Glenny AM, Worthington HV, Littlewood A, Fernandez Mauleffinch LM, Clarkson JE and McCabe MG: Interventions for preventing oral mucositis in patients with cancer receiving treatment: Cytokines and growth factors. Cochrane Database Syst Rev. 11:CD0119902017.PubMed/NCBI

130 

Esquivel-Velázquez M, Ostoa-Saloma P, Palacios-Arreola MI, Nava-Castro KE, Castro JI and Morales-Montor J: The role of cytokines in breast cancer development and progression. J Interferon Cytokine Res. 35:1–16. 2015. View Article : Google Scholar : PubMed/NCBI

131 

Hosokawa H and Rothenberg EV: Cytokines, transcription factors, and the initiation of T-cell development. Cold Spring Harb Perspect Biol. 10:a0286212018. View Article : Google Scholar : PubMed/NCBI

132 

Dayakar A, Chandrasekaran S, Kuchipudi SV and Kalangi SK: Cytokines: Key determinants of resistance or disease progression in visceral leishmaniasis: Opportunities for novel diagnostics and immunotherapy. Front Immunol. 10:6702019. View Article : Google Scholar : PubMed/NCBI

133 

Wang X, Wong K, Ouyang W and Rutz S: Targeting IL-10 family cytokines for the treatment of human diseases. Cold Spring Harb Perspect Biol. 11:a0285482019. View Article : Google Scholar : PubMed/NCBI

134 

Ali MF, Driscoll CB, Walters PR, Limper AH and Carmona EM: β-Glucan-activated human B lymphocytes participate in innate immune responses by releasing proinflammatory cytokines and stimulating neutrophil chemotaxis. J Immunol. 195:5318–5326. 2015. View Article : Google Scholar : PubMed/NCBI

135 

Trottein F and Paget C: Natural killer T cells and mucosal-associated invariant T cells in lung infections. Front Immunol. 9:17502018. View Article : Google Scholar : PubMed/NCBI

136 

Isaeva OI, Sharonov GV, Serebrovskaya EO, Turchaninova MA, Zaretsky AR, Shugay M and Chudakov DM: Intratumoral immunoglobulin isotypes predict survival in lung adenocarcinoma subtypes. J Immunother Cancer. 7:2792019. View Article : Google Scholar : PubMed/NCBI

137 

Fajgenbaum DC and June CH: Cytokine storm. N Engl J Med. 383:2255–2273. 2020. View Article : Google Scholar : PubMed/NCBI

138 

Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, Herridge M, Randolph AG and Calfee CS: Acute respiratory distress syndrome. Nat Rev Dis Primers. 5:182019. View Article : Google Scholar : PubMed/NCBI

139 

Eskilsson A, Shionoya K, Engblom D and Blomqvist A: Fever during localized inflammation in mice is elicited by a humoral pathway and depends on brain endothelial interleukin-1 and interleukin-6 signaling and central EP3 receptors. J Neurosci. 41:5206–5218. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Kang S and Kishimoto T: Interplay between interleukin-6 signaling and the vascular endothelium in cytokine storms. Exp Mol Med. 53:1116–1123. 2021. View Article : Google Scholar : PubMed/NCBI

141 

Tang XD, Ji TT, Dong JR, Feng H, Chen FQ, Chen X, Zhao HY, Chen DK and Ma WT: Pathogenesis and treatment of cytokine storm induced by infectious diseases. Int J Mol Sci. 22:130092021. View Article : Google Scholar : PubMed/NCBI

142 

Hemminki O, Dos Santos JM and Hemminki A: Oncolytic viruses for cancer immunotherapy. J Hematol Oncol. 13:842020. View Article : Google Scholar : PubMed/NCBI

143 

Kaufman HL, Kohlhapp FJ and Zloza A: Oncolytic viruses: A new class of immunotherapy drugs. Nat Rev Drug Discov. 14:642–646. 2015. View Article : Google Scholar : PubMed/NCBI

144 

Rahman MM and McFadden G: Oncolytic viruses: Newest frontier for cancer immunotherapy. Cancers (Basel). 13:54522021. View Article : Google Scholar : PubMed/NCBI

145 

Innao V, Rizzo V, Allegra AG, Musolino C and Allegra A: Oncolytic viruses and hematological malignancies: A new class of immunotherapy drugs. Curr Oncol. 28:159–183. 2020. View Article : Google Scholar : PubMed/NCBI

146 

Heidbuechel JPW and Engeland CE: Oncolytic viruses encoding bispecific T cell engagers: A blueprint for emerging immunovirotherapies. J Hematol Oncol. 14:632021. View Article : Google Scholar : PubMed/NCBI

147 

Liu S, Liu F, Zhao M and Zhang J: Antitumor efficacy of oncolytic herpes virus type 1 armed with GM-CSF in murine uveal melanoma xenografts. Cancer Manag Res. 12:11803–11812. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Lühl NC, Zirngibl F, Dorneburg C, Wei J, Dahlhaus M, Barth TF, Meyer LH, Queudeville M, Eckhoff S, Debatin KM and Beltinger C: Attenuated measles virus controls pediatric acute B-lineage lymphoblastic leukemia in NOD/SCID mice. Haematologica. 99:1050–1061. 2014. View Article : Google Scholar : PubMed/NCBI

149 

Ammayappan A, Russell SJ and Federspiel MJ: Recombinant mumps virus as a cancer therapeutic agent. Mol Ther Oncolytics. 3:160192016. View Article : Google Scholar : PubMed/NCBI

150 

Bommareddy PK, Patel A, Hossain S and Kaufman HL: Talimogene laherparepvec (T-VEC) and other oncolytic viruses for the treatment of melanoma. Am J Clin Dermatol. 18:1–15. 2017. View Article : Google Scholar : PubMed/NCBI

151 

Zhang R, Cui Y, Guan X and Jiang X: A recombinant human adenovirus type 5 (H101) combined with chemotherapy for advanced gastric carcinoma: A retrospective cohort study. Front Oncol. 11:7525042021. View Article : Google Scholar : PubMed/NCBI

152 

Omole RK, Oluwatola O, Akere MT, Eniafe J, Agboluaje EO, Daramola OB, Ayantunji YJ, Omotade TI, Torimiro N, Ayilara MS, et al: Comprehensive assessment on the applications of oncolytic viruses for cancer immunotherapy. Front Pharmacol. 13:10827972022. View Article : Google Scholar : PubMed/NCBI

153 

Mardi A, Shirokova AV, Mohammed RN, Keshavarz A, Zekiy AO, Thangavelu L, Mohamad TAM, Marofi F, Shomali N, Zamani A and Akbari M: Biological causes of immunogenic cancer cell death (ICD) and anti-tumor therapy; combination of oncolytic virus-based immunotherapy and CAR T-cell therapy for ICD induction. Cancer Cell Int. 22:1682022. View Article : Google Scholar : PubMed/NCBI

154 

Ajina A and Maher J: Prospects for combined use of oncolytic viruses and CAR T-cells. J Immunother Cancer. 5:902017. View Article : Google Scholar : PubMed/NCBI

155 

Spiesschaert B, Angerer K, Park J and Wollmann G: Combining oncolytic viruses and small molecule therapeutics: Mutual benefits. Cancers (Basel). 13:33862021. View Article : Google Scholar : PubMed/NCBI

156 

Schirrmacher V: Molecular mechanisms of anti-neoplastic and immune stimulatory properties of oncolytic Newcastle disease virus. Biomedicines. 10:5622022. View Article : Google Scholar : PubMed/NCBI

157 

Globerson-Levin A, Waks T and Eshhar Z: Elimination of progressive mammary cancer by repeated administrations of chimeric antigen receptor-modified T cells. Mol Ther. 22:1029–1038. 2014. View Article : Google Scholar : PubMed/NCBI

158 

Chang YH, Lau KS, Kuo RL and Horng JT: dsRNA binding domain of PKR is proteolytically released by enterovirus A71 to facilitate viral replication. Front Cell Infect Microbiol. 7:2842017. View Article : Google Scholar : PubMed/NCBI

159 

Zhang X, Wang S, Zhu Y, Zhang M, Zhao Y, Yan Z, Wang Q and Li X: Double-edged effects of interferons on the regulation of cancer-immunity cycle. Oncoimmunology. 10:19290052021. View Article : Google Scholar : PubMed/NCBI

160 

Ginting TE, Christian S, Larasati YO, Suryatenggara J, Suriapranata IM and Mathew G: Antiviral interferons induced by Newcastle disease virus (NDV) drive a tumor-selective apoptosis. Sci Rep. 9:151602019. View Article : Google Scholar : PubMed/NCBI

161 

Roulstone V, Mansfield D, Harris RJ, Twigger K, White C, de Bono J, Spicer J, Karagiannis SN, Vile R, Pandha H, et al: Antiviral antibody responses to systemic administration of an oncolytic RNA virus: the impact of standard concomitant anticancer chemotherapies. J Immunother Cancer. 9:e0026732021. View Article : Google Scholar : PubMed/NCBI

162 

Cheng JT, Wang YY, Zhu LZ, Zhang Y, Cai WQ, Han ZW, Zhou Y, Wang XW, Peng XC, Xiang Y, et al: Novel transcription regulatory sequences and factors of the immune evasion protein ICP47 (US12) of herpes simplex viruses. Virol J. 17:1012020. View Article : Google Scholar : PubMed/NCBI

163 

Wang G, Kang X, Chen KS, Jehng T, Jones L, Chen J, Huang XF and Chen SY: An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor neoantigen-specific T cell responses. Nat Commun. 11:13952020. View Article : Google Scholar : PubMed/NCBI

164 

Arnone CM, Polito VA, Mastronuzzi A, Carai A, Diomedi FC, Antonucci L, Petrilli LL, Vinci M, Ferrari F, Salviato E, et al: Oncolytic adenovirus and gene therapy with EphA2-BiTE for the treatment of pediatric high-grade gliomas. J Immunother Cancer. 9:e0019302021. View Article : Google Scholar : PubMed/NCBI

165 

Shi T, Song X, Wang Y, Liu F and Wei J: Combining oncolytic viruses with cancer immunotherapy: Establishing a new generation of cancer treatment. Front Immunol. 11:6832020. View Article : Google Scholar : PubMed/NCBI

166 

Guedan S and Alemany R: CAR-T cells and oncolytic viruses: Joining forces to overcome the solid tumor challenge. Front Immunol. 9:24602018. View Article : Google Scholar : PubMed/NCBI

167 

Chianese A, Santella B, Ambrosino A, Stelitano D, Rinaldi L, Galdiero M, Zannella C and Franci G: Oncolytic viruses in combination therapeutic approaches with epigenetic modulators: Past, present, and future perspectives. Cancers (Basel). 13:27612021. View Article : Google Scholar : PubMed/NCBI

168 

Luo Q, Pan M, Feng H and Wang L: ABO blood group antigen therapy: A potential new strategy against solid tumors. Sci Rep. 11:162412021. View Article : Google Scholar : PubMed/NCBI

169 

O'Brien RM, Cannon A, Reynolds JV, Lysaght J and Lynam-Lennon N: Complement in tumourigenesis and the response to cancer therapy. Cancers (Basel). 13:12092021. View Article : Google Scholar : PubMed/NCBI

170 

Khalili H, Wolpin BM, Huang ES, Giovannucci EL, Kraft P, Fuchs CS and Chan AT: ABO blood group and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev. 20:1017–1020. 2011. View Article : Google Scholar : PubMed/NCBI

171 

Akin S and Altundag K: Clinical associations with ABO blood group and rhesus blood group status in patients with breast cancer: A nationwide retrospective study of 3,944 breast cancer patients in Turkey. Med Sci Monit. 24:4698–4703. 2018. View Article : Google Scholar : PubMed/NCBI

172 

Elkoshi Z: Cancer and autoimmune diseases: A tale of two immunological opposites? Front Immunol. 13:8215982022. View Article : Google Scholar : PubMed/NCBI

173 

Kolev M and Markiewski MM: Targeting complement-mediated immunoregulation for cancer immunotherapy. Semin Immunol. 37:85–97. 2018. View Article : Google Scholar : PubMed/NCBI

174 

Makunts T, Saunders IM, Cohen IV, Li M, Moumedjian T, Issa MA, Burkhart K, Lee P, Patel SP and Abagyan R: Myocarditis occurrence with cancer immunotherapy across indications in clinical trial and post-marketing data. Sci Rep. 11:173242021. View Article : Google Scholar : PubMed/NCBI

175 

Ermer T, Canavan ME, Maduka RC, Li AX, Salazar MC, Kaminski MF, Pichert MD, Zhan PL, Mase V, Kluger H and Boffa DJ: Association between food and drug administration approval and disparities in immunotherapy use among patients with cancer in the US. JAMA Netw Open. 5:e22195352022. View Article : Google Scholar : PubMed/NCBI

176 

Noman MZ, Hasmim M, Lequeux A, Xiao M, Duhem C, Chouaib S, Berchem G and Janji B: Improving cancer immunotherapy by targeting the hypoxic tumor microenvironment: New opportunities and challenges. Cells. 8:10832019. View Article : Google Scholar : PubMed/NCBI

177 

Kawashima S, Inozume T, Kawazu M, Ueno T, Nagasaki J, Tanji E, Honobe A, Ohnuma T, Kawamura T, Umeda Y, et al: TIGIT/CD155 axis mediates resistance to immunotherapy in patients with melanoma with the inflamed tumor microenvironment. J Immunother Cancer. 9:e0031342021. View Article : Google Scholar : PubMed/NCBI

178 

Ma Z, Zhang W, Dong B, Xin Z, Ji Y, Su R, Shen K, Pan J, Wang Q and Xue W: Docetaxel remodels prostate cancer immune microenvironment and enhances checkpoint inhibitor-based immunotherapy. Theranostics. 12:4965–4979. 2022. View Article : Google Scholar : PubMed/NCBI

179 

Lee HT, Lee SH and Heo YS: Molecular interactions of antibody drugs targeting PD-1, PD-L1, and CTLA-4 in immuno-oncology. Molecules. 24:11902019. View Article : Google Scholar : PubMed/NCBI

180 

Capuano C, Pighi C, Battella S, De Federicis D, Galandrini R and Palmieri G: Harnessing CD16-mediated NK cell functions to enhance therapeutic efficacy of tumor-targeting mAbs. Cancers (Basel). 13:25002021. View Article : Google Scholar : PubMed/NCBI

181 

Li Q, Oduro PK, Guo R, Li R, Leng L, Kong X, Wang Q and Yang L: Oncolytic viruses: Immunotherapy drugs for gastrointestinal malignant tumors. Front Cell Infect Microbiol. 12:9215342022. View Article : Google Scholar : PubMed/NCBI

182 

Burn OK, Prasit KK and Hermans IF: Modulating the tumour microenvironment by intratumoural injection of pattern recognition receptor agonists. Cancers (Basel). 12:38242020. View Article : Google Scholar : PubMed/NCBI

183 

Couzin-Frankel J: Breakthrough of the year 2013. Cancer immunotherapy. Science. 342:1432–1433. 2013. View Article : Google Scholar : PubMed/NCBI

184 

Liu K, Cui JJ, Zhan Y, Ouyang QY, Lu QS, Yang DH, Li XP and Yin JY: Reprogramming the tumor microenvironment by genome editing for precision cancer therapy. Mol Cancer. 21:982022. View Article : Google Scholar : PubMed/NCBI

185 

Sharma VP, Tang B, Wang Y, Duran CL, Karagiannis GS, Xue EA, Entenberg D, Borriello L, Coste A, Eddy RJ, et al: Live tumor imaging shows macrophage induction and TMEM-mediated enrichment of cancer stem cells during metastatic dissemination. Nat Commun. 12:73002021. View Article : Google Scholar : PubMed/NCBI

186 

Virant-Klun I, Skerl P, Novakovic S, Vrtacnik-Bokal E and Smrkolj S: Similar population of CD133+ and DDX4+ VSEL-like stem cells sorted from human embryonic stem cell, ovarian, and ovarian cancer ascites cell cultures: The real embryonic stem cells? Cells. 8:7062019. View Article : Google Scholar : PubMed/NCBI

187 

Yan Y, Chen Y, Yang F, Chen IH, Xiong Z, Wang J, Lachman LB, Wang H and Yang XF: HLA-A2.1-restricted T cells react to SEREX-defined tumor antigen CML66L and are suppressed by CD4+CD25+ regulatory T cells. Int J Immunopathol Pharmacol. 20:75–89. 2007. View Article : Google Scholar : PubMed/NCBI

188 

Wargo JA, Robbins PF, Li Y, Zhao Y, El-Gamil M, Caragacianu D, Zheng Z, Hong JA, Downey S, Schrump DS, et al: Recognition of NY-ESO-1+ tumor cells by engineered lymphocytes is enhanced by improved vector design and epigenetic modulation of tumor antigen expression. Cancer Immunol Immunother. 58:383–394. 2009. View Article : Google Scholar : PubMed/NCBI

189 

Yang L, Gu X, Yu J, Ge S and Fan X: Oncolytic virotherapy: From bench to bedside. Front Cell Dev Biol. 9:7901502021. View Article : Google Scholar : PubMed/NCBI

190 

Han W, Li W, Zhang X, Du Z, Liu X, Zhao X, Wen X, Wang G, Hu JF and Cui J: Targeted breast cancer therapy by harnessing the inherent blood group antigen immune system. Oncotarget. 8:15034–15046. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2023
Volume 49 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao F, Shen D, Shang M, Yu H, Zuo X, Chen L, Huang Z, Li L and Wang L: Immunotherapy: A new target for cancer cure (Review). Oncol Rep 49: 100, 2023
APA
Zhao, F., Shen, D., Shang, M., Yu, H., Zuo, X., Chen, L. ... Wang, L. (2023). Immunotherapy: A new target for cancer cure (Review). Oncology Reports, 49, 100. https://doi.org/10.3892/or.2023.8537
MLA
Zhao, F., Shen, D., Shang, M., Yu, H., Zuo, X., Chen, L., Huang, Z., Li, L., Wang, L."Immunotherapy: A new target for cancer cure (Review)". Oncology Reports 49.5 (2023): 100.
Chicago
Zhao, F., Shen, D., Shang, M., Yu, H., Zuo, X., Chen, L., Huang, Z., Li, L., Wang, L."Immunotherapy: A new target for cancer cure (Review)". Oncology Reports 49, no. 5 (2023): 100. https://doi.org/10.3892/or.2023.8537