Next Article in Journal
Bioinformatic Analysis and In Vitro and In Vivo Experiments Reveal That Fibrillarin Participates in the Promotion of Lung Metastasis in Hepatocellular Carcinoma
Previous Article in Journal
Corneal Adhesion Possesses the Characteristics of Solid and Membrane
Previous Article in Special Issue
Emerging Promise of Computational Techniques in Anti-Cancer Research: At a Glance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Tumor Effect of Parasitic Protozoans

1
School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310059, China
2
Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
3
College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou 311300, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Bioengineering 2022, 9(8), 395; https://doi.org/10.3390/bioengineering9080395
Submission received: 21 July 2022 / Revised: 12 August 2022 / Accepted: 12 August 2022 / Published: 16 August 2022
(This article belongs to the Special Issue Targeted Therapy for Cancer)

Abstract

:
The immune system may aberrantly silence when against “altered self”, which consequently may develop into malignancies. With the development of tumor immunology and molecular biology, the deepened understanding of the relationship between parasites and tumors shifts the attitude towards parasitic pathogens from elimination to utilization. In recent years, the antitumor impact implemented by protozoan parasites and the derived products has been confirmed. The immune system is activated and enhanced by some protozoan parasites, thereby inhibiting tumor growth, angiogenesis, and metastasis in many animal models. In this work, we reviewed the available information on the antitumor effect of parasitic infection or induced by parasitic antigen, as well as the involved immune mechanisms that modulate cancer progression. Despite the fact that clinical trials of the protozoan parasites against tumors are limited and the specific mechanisms of the effect on tumors are not totally clear, the use of genetically modified protozoan parasites and derived molecules combined with chemotherapy could be an important element for promoting antitumor treatment in the future.

Graphical Abstract

1. Introduction

Cancer is a growing global disease with no borders. It is regarded as one of the leading causes of mortality in humans, with an estimated 19.3 million new cancer cases and almost 10.0 million cancer deaths occurring in 2020, according to Global Cancer Statistics [1]. The development of efficient therapeutic strategies is important. In addition to traditional cancer treatments including surgery, radiotherapy, and chemotherapy, more recently, cancer biotherapies such as cancer vaccines, monoclonal antibodies, gene therapy drugs, and immunomodulators have been used clinically and studied in clinical trials. By killing cancer cells or inhibiting their growth, the mechanism of biotherapy mainly induces defensive immune responses against tumors in multiple targets and directions, in contrast to traditional treatment [2]. In recent years, some parasites, especially intracellular protozoan parasites, have been considered effectors to induce anti-pathogen and anti-tumor immune responses so as to overcome tumor escape and active tumor surveillance system. The persistent antigen release caused by the chronic infection of protozoan parasites leads to a long-term specific immune response, which may have broader advantages for promoting cancer treatments. This review outlined the mechanism of the anti-tumor effect of parasitic protozoon, aiming to provide novel strategies for the clinical treatment of tumors.

2. Tumor Therapy with the Injection of Parasite

Recently, the use of non-pathogenic live protozoan parasites as anticancer therapeutic approaches has drawn attention worldwide, of note is the long history of antitumor effect by Trypanosoma cruzi. At the very beginning in 1946, the work from the former Soviet Union reported that cancer is rare in patients infected with T. cruzi once before [3], which opened possibilities for research on cancer biotherapy. Later, researchers developed an anticancer experiment in which T. cruzi extracts were directly inoculated in peritumoral areas over different tumors, and all of the results showed a reduction in tumor size [4]. However, the work was hampered by controversial results and the complicated international political situation, and so the molecular basis of this phenomenon has remained elusive [5]. Moreover, several live-attenuated protozoan parasites including Leishmania infantum and L. tropica [6], Neospora caninum [7], and Toxoplasma gondii [8] have all lately been employed as antitumor biological agents via intratumoral injection, all of which decreased tumor development and local inflammation (Figure 1). The injection of live N. caninum tachyzoites either in or remotely from the tumor, successfully treated murine thymoma EG7 by strongly activating the natural killer cell- (NK cell-) and CD8-T cell-dependent protective antitumor response associated with interferon (IFN)-γ secretion in the tumor microenvironment, resulting in the lysis of the cancer cell [7]. Researchers from Ege University Medical School first reported that intratumoral administration of attenuated Leishmania strains in 4T1 breast cancer-bearing mice promoted M1 dominant activation of macrophages in spleen and tumor tissues with induction of proinflammatory cytokines that helps the generation of protective Th1 responses [6,9]. It is a pity that the survival rate and other antitumor effects were not recorded. Further studies on the molecular basis and the effect on different tumors needs to be carried out to deepen the understanding and application of antitumor biotherapy with Leishmania strains.
The parasite T. gondii, on the other hand, attracts more attention. The characteristic of T. gondii that can infect nearly all types of cells and modify the immune response of the host opens up a wide range of clinical possibilities for T. gondii as an oncolytic protozoan in human medicine [10]. Considering that the toxoplasma virulence is cell type-independent, researchers in 1985 performed an intralesional injection with 107 formalin-fixed Toxoplasma tachyzoites one day after the syngeneic Lewis lung carcinoma cell inoculation and discovered some mice (2/6) completely rejected the growth of the implanted tumor [11]. It is conceivable that live organisms could elicit a much more robust antitumor effect than dead ones. Thus, what followed was gamma radiation-attenuated Toxoplasma [12], and gene-edited-attenuated Toxoplasma such as ΔCPS, ΔOMPDCΔUP, ΔGRA17 strains, and Δldh1-Δldh2 strains used for tumor biotherapy, which all presented the ability to repress the growth of established tumors and to help the inhibition of lethal tumor development in the mice [8,13,14]. Overall, these protozoan parasites are proven to have effective antitumor effects, while the precise targets of tumor cells need further investigation. Referred to as the chimeric antigen receptor (CAR)-T method, whether the combination of protozoan parasites and tumor-specific antigens would promote the target invasion and antitumor effects requires further research data.

3. Antitumor Effect of Parasitic Products

Some parasitic products have been demonstrated to have specific antitumor effects. It is well known that the levels of aberrant chondroitin sulfate proteoglycans (CSPGs), a protein family that displays one or multiple chondroitin sulfate (CS) side chains, are upregulated in many cancers but the variability of this protein within different tumor tissues is striking, due to the great diversity of structure cores assembled by numerous enzymes regulated, based on tissue and cell type [15,16]. The CS side chains or the protein core of CSPGs can bind extracellular matrix components or a growth factor receptor complex to transmit pro-oncogenic signaling through which the proteins are implicated in cancers. The identification and targeting of CS in cancers remained a technical challenge until the specific CS structure, termed oncofetal CS (ofCS) was found to share a high affinity among cancer cells [17]. A refined malaria protein called rVAR2 was discovered to bind with the distinct ofCS from a variety of different cancer cell lines [18], with remarkably high specificity and affinity (KD ~15 nM) [19]. This discovery was further supported by the observation that rVAR2 intravenously injected, adhered to the tumors in in vivo xenograft animal models [20]. The high efficiency targeted tumor characteristic of rVAR2 makes it a potentially ideal carrier for anti-cancer drug delivery. Indeed, it has been proven that a hemiasterlin analog (KT886) conjugated rVAR2 (VDC886), formed as the complex which carried an average of three toxins per rVAR2 molecule, effectively kill a total of 33 cancer cell lines in vitro with inhibitory concentration 50 (IC50) values ranging from 0.2 pM to 30 nM [18]. Remarkably, the VDC886 treatment significantly slowed the growth of both non-Hodgkin’s lymphoma (Karpas299) and prostate cancer (PC-3) in mouse models, indicating a potential role in clinic therapy.
Unlike malaria VAR2, rhoptry and dense granule proteins secreted by T. gondii play advantageous roles in regulating the growth of tumors. Two key organelles, the apical rhoptry [21] and dense granule [22] are essential for host invasion and immune escape. Through a reverse genetic approach to target complete gene deletions, particular proteins including rhoptry protein 5 (ROP5), ROP17, ROP18, ROP35, and ROP38, the dense granule protein 2 (GRA2), GRA12, and GRA24 are demonstrated to effectively activate antitumor immune responses involving CD4+ and CD8+ T cells and the interleukin-12 (IL-12)/IFN-γ TH1 axis, while deletion of GRA3, GRA15, GRA16, ROP16, or ROP21 did not affect the antitumor activity [23]. Coincidentally, the secreted GRA15 was confirmed to localize to the endoplasmic reticulum and to activate innate immune and stimulator of interferon genes (STING) responses by promoting polyubiquitination at Lys-337 and oligomerization in a tumor necrosis factor (TNF) receptor-associated factor (TRAF) protein-dependent manner [24]. In addition, the GRA16 displayed the ability to break the chemoresistance of irinotecan by inhibiting nuclear factor kappa B (NF-κB) via a PP2A-B55/AKT/NF-κB p65 pathway against non-small-cell lung carcinoma (NSCLC) [25]. Recently, the fact that the small intestine (SI) exhibited low tumorigenesis or metastatic growth from distant tumors attracted attention. Concomitantly, a protein named Eimeria antigen (EA) that acts as a robust stimulator to promote IL-12 releasing from dendritic cells (DC) and to upregulating inflammatory modulators (Monocyte chemoattractant protein-1 (MCP-1), IL-6, IFN-γ, and TNF-α) for the defending of sarcoma tumor in mice was discovered [26]. Perhaps the vast immunologic compartment induced by EA in SI also provides a potent tumor immunosurveillance. These findings collectively point to a deeper understanding of fundamental mechanisms of host cell pathways manipulated by these secreted proteins, which could provide novel targets for developing effective therapies against aggressive solid tumors.
Another well-known anti-tumor protein is the calreticulin from T. cruzi (TcCRT), which is secreted into the extracellular milieu to induce immune modulations [27,28]. TcCRT consists of an N-terminal vasostatin-like domain (aa 20-193) that can directly bind with endothelial cells through a scavenger-like receptor and acts as a potent angiogenesis inhibitor [29,30]. As we all know, angiogenesis-focused therapy is frequently utilized in conjunction with other treatments for a variety of tumor types [31,32]. Most likely, by directly interacting with endothelial cells, the N-terminal vasostatin-like domain inhibits vascular endothelial growth factor (VEGF)-induced cell proliferation and induces cell apoptosis [33]. TcCRT makes use of its anti-tumor properties in this manner.

4. Activating the Cellular Immune System

Antitumor immunity including innate and adaptive immune responses, mainly activated depending on the close interaction with several elements, has the most important role in tumor control [34]. However, tumor cells often evolve many mechanisms to escape immune surveillance and elimination [35]. Therefore, disrupting these mechanisms and promoting tumor cell recognition has the potential to effectively boost anticancer immunity. In some way, some protozoan parasites and their products may facilitate targeting tumor cells and induce positive immune responses, yielding long-term benefits of immunity against tumors, such as immune cells and potent cytokines.

4.1. Immune Cells

Cells of innate immunity and adaptive immunity, including NK cells, DC, Innate lymphoid cells (ILC), cytokine-induced killer cells (LAK), specific T lymphocytes, etc., play important roles in tumor immunotherapy by controlling tumor growth and killing tumor cells.
Acting as toxic immune cells, activated NK cells managed by a suite of activating, co-stimulatory and inhibitory receptors [36], can directly kill tumor cells, especially those that lack major histocompatibility complex (MHC; also known as human leukocyte antigen (HLA)) class I [37], such as tumor cells from metastatic and blood tumors, and indirectly improve the response of antibodies and T cells [38]. By the stimulatory infection of Plasmodium, NK cells can be activated to kill some lung cancer cells which in return can release tumor antigens resulting in activation of the systemic response of tumor antigen-specific T cells in peripheral blood, spleen, and lymph nodes, as the consequence of additional cancer cell death [39]. Possibly, the toxic activation of NK cells via IFN-γ expression is induced by the infection of Plasmodium through the up-regulated expression of CD69 and CD25 [40]. As reported by Chen, et al. [41], three clinical trials based on Plasmodium immunotherapy against advanced cancers have been approved and are underway with clinical safety guaranteed. In addition, the infection of N. caninum [7], Leishmania amazonensis [42], and T. gondii [43] increased the levels of NK cells mainly through an IFN-γ-dependent pathway. Moreover, toxoplasma infection can induce the conversion of NK cells into ILC1-like cells which are Eomes-dependent and the changes appear permanent [44], which may help defend against tumors. However, the molecular mechanism of how these protozoans activate NK cells is not well studied. While the role of NK cells in controlling tumor growth is well established, the function of newly discovered ILCs in defending tumors remains poorly understood. Nevertheless, their ability to produce large amounts of cytokines [45,46] indicates that ILCs may contribute to tumor-associated inflammation, and the interaction between protozoan parasites and ILCs may shed light on the development of new antitumor therapies. Genetic modification with superior activation or decreased inhibitory signals in NK cells enhances their tumor cell killing ability [47]. It is conceivable when using the specific parasitic molecular, such as VAR2 form Plasmodium, as the chimeric antigen receptor (CAR) in NK cells, the most effective CARs used to redirect T cells also work well for NK cells [48,49].
Undoubtedly, T cells play crucial roles against tumors [50]. Activation of naïve T lymphocytes requires T cell receptor (TCR) signaling, costimulatory signaling, and cytokine support [51,52]. A neoantigen peptide, uniquely encoded by mutated DNA of tumor cells, has distinct epitopes from those of normal cells and will be processed and displayed on the surface of tumor cells and antigen-presenting cells (APC) as the form of neoantigen peptide-major histocompatibility complexes (pMHC) [53,54]. During T cell activation, the expressed membrane proteins CD4 on T helper cells and CD8 on cytotoxic T lymphocytes, bind to MHC class II (MHC-II) or MHC-I molecules, respectively [55]. However, recent evidence revealed that the multifaceted suppressive signals resulted in an exhausted phenotype or dysfunctional state of T cells [56,57]. Hence, effective therapeutic immunity against tumors can be stimulated by reversing tumor-associated immunosuppression. So far, a few studies have indicated that this goal can be addressed using the infection of protozoans, which may reverse T cell suppression and activation by providing exogenous antigens. Mice immunized with live-attenuated Leishmania parasites presented a higher percentage of CD4+ and CD8+ T cells, indicating a robust cellular response was generated [58,59], consequently these T cells infiltrated the 4T1 breast cancer resulting in the decrease of tumor volume and prolonged the survival period of mice [6]. Similarly, vaccination with T. cruzi significantly inhibited the growth of breast and colon tumors through the activation of CD4+ and CD8+ T cells and the increase of macrophages and dendritic cells. A diverse cell-mediated (CD4+ and CD8+ T cells) immunity lasted more than 200 days in mice that were triggered by an attenuated T. gondii against pancreatic cancer recurrences [60].
Although DC cannot directly kill tumor cells [61], they are the cornerstone of the anti-tumor immune response due to the ability to activate T cells by extracting and transporting specific tumor antigens making them the foundation of the anti-tumor immune response [62]. However, it is usually insufficient for DC maturation due to the suppressive mechanisms within tumors [63], including suppressing the expression of MHC or costimulatory molecules [64]. Some protozoan parasites may help promote the mutation of DCs, and this notion is supported by studies demonstrating that ligand molecules from parasites bind with Toll-like receptors (TLRs) to stimulate DC activation, such as glycosylphosphatidylinositol (GPI) from Leishmania major [65], T. cruzi [66], P. falciparum [67], and T. gondii [68], and Profilin-like protein from T. gondii [69]. In mice implanted with murine Lewis lung cancer (LLC) cells, malaria parasite infection promotes the maturation of DCs through up-regulating the expression of CD80 and CD86 resulting in the activation of T cells [39]. Recently, Lantier, et al. [7] found that unlike dead parasites or soluble tachyzoites antigens, live N. caninum tachyzoites are able to activate murine or human DCs to secret proinflammatory cytokines, which convincingly suggests that the use of live tachyzoites is a necessary condition for immunotherapeutic treatment. In fact, four days after the injection of live tachyzoites, they observed recruitment of DCs, along with a high increase of IFN-γ and IL-12, indicating a measurable and systemic immune response against EG7 thymoma in mice. Coincidentally, Baird, et al. [14] found that CD11c+ DC antigen-presenting cells from the ovarian carcinoma microenvironment invaded by the infection of toxoplasma cps strain, strongly upregulate the costimulatory molecules CD80 and CD86, which regained the ability to cross-present antigen to prime tumor antigen-specific CD8+ T cell responses.

4.2. Cytokines

Cytokines are major regulators of innate and adaptive immunity, and some play critical roles in tumor cells [70]. Among these molecules, IFN-γ can induce tumor cell cycle arrest and establish tumor cell dormancy [71], and IL-12 can promote Th1 antitumor immune response which may be secreted by the stimulation of IFN-γ and in turn triggers the re-activation of the IFN-γ production cycle [72], while IL-10 and IL-13 can inhibit Th1 cells from secreting IFN-γ [73]. The highly expressed IL-6 can promote inflammation and tumor cell immunosuppression [74], and possibly aggravate tumor cell growth and metastasis by IL-6/Signal Transducer And Activator Of Transcription 3 (STAT3) mediated inhibition of DC and lead to the dysfunction of the immune system [75]. Generally speaking, cytokines dominated by IL-6 are beneficial for tumor proliferation and metastasis, while these inflammation factors mediated by IFN-γ have an anti-tumor effect on blocking tumor progression [76]. For instance, exposure to Toxoplasma tachyzoites, in addition to activating immune cells as mentioned above, can induce the antitumor effect in mice models by increasing the expression levels of IFN-γ [8,13,23,43,77], IL-12 [14,23,43,78], and TNF-α [8,13].

5. Activating the Humoral Immunity System

A long-term, effective anti-tumor immune response is essential for the treatment of tumors, and during this period, some tumor-associated antigens (TAA), which are at very low levels in normal cells, will be recognized and prime the humoral immunity [79]. The auto-tolerance prevents the direct recognition against self-antigen, but the infection of protozoan may help induce the anti-tumor humoral immunity followed by the releasing of antibodies which can affect the biology of the tumor by blocking certain receptors on the surface of tumor cells [80]. Infection of toxoplasma ME49 strains significantly increase the levels of IgG1 and IgG2a in LLC-bearing mice [77], and, live L. tarentolae carrying E7 protein induced significant levels of IgG2a against HPV-associated tumors [81]. It would be more convincing if the tumor-specific IgG was detected. The similarity between surface antigens of T. cruzi and Ehrlich’s adenocarcinoma cells was confirmed by the cross-reaction of indirect immunofluorescence [82]. Later, the sera from T. cruzi lysate-vaccinated mice significantly reduced the tumor size of Ehrlich’s adenocarcinoma. This could imply that the specific immune profile generated by antigens of T. cruzi has a positive effect on the growth of a tumor, at least regarding this type of neoplasm.

6. Suppressing the Angiogenesis and Tumor Metastasis

Angiogenesis, the process of recruiting new blood vessels, is an essential component of tumor metastasis to acquire adequate nutrients and oxygen to facilitate the spread and growth of tumor cells. Among the proangiogenic molecules, the acidic and basic fibroblast growth factors (aFGF and bFGF), the platelet-derived endothelial cell growth factor (PD-ECGF), and especially the VEGF are well studied [83]. Attenuated T. gondii ME49 remarkably suppressed the levels of angiogenic factors (VEGF, integrin, matrix metallopeptidase (MMP) 2, and MMP9) in an Ehrlich ascites carcinoma cancer model, which in turn inhibited neoplastic growth [12]. It is interesting to note that protozoan parasites suppress neovascularization through quite different pathways. For instance, the infection of Plasmodium exhibited an inhibitory effect on hepatocellular carcinoma angiogenesis, unlike the effect of T. gondii mentioned above, further detection showed no significant decrease in VEGF levels [84]. According to many reports, VEGF is known as a major proangiogenic molecular released by tumor-associated macrophages (TAMs) and is correlated with many human cancers [85]. Later, they discovered the diminished angiogenic responses in Plasmodium-infected tumor-bearing mice probably by decreasing the expression of angiogenesis-related enzyme MMP9 via the insulin-like growth factor (IGF) axis through the PI3-K and MAPK signaling pathways [41,84]. In addition, the parasite-derived antigens have an effort on inhibiting vascularization in tumors as well. It has been reported that T. cruzi CRT or more precisely, its N-terminus, directly interacts with endothelial cells through VEGF and inhibits angiogenesis in breast tumors in vitro, ex vivo, and in vivo [86,87]. In the treatment of mice with T. gondii lysate antigen (TLA) in the murine sarcoma-180 tumors model, both the decreased expression of CD31 (an angiogenesis marker in the tumor tissue) and the reduction in tumor size were observed [88].

7. Improving the Microenvironment

Cancer immunotherapy based on immune checkpoint blockade has shown its value in clinical treatments, but unfortunately, only a small number of tumor-bearing patients benefit from the therapy. The failure is likely due to the low immunogenicity, high numbers of immunosuppressive cells, and insufficient T cell infiltration in the tumor microenvironment (TME) [89]. Myeloid-derived immunosuppressive cells (MDSCs), a group of immature myeloid cells at different development stages, can suppress anti-cancer immune responses by inhibiting the antitumor effects of CD8+ T cells [90]. Regulator T cells (Tregs) are a subset of immunosuppressive T cells that are recruited into the tumor microenvironment by interacting with various tumor-secreted chemokines [91]. MDSCs and Tregs are believed to play a major role in the suppression of antitumor immune responses [92,93]. There are some reports that Plasmodium and T. gondii can reduce the number of immunosuppressive cells (Figure 2). For instance, in the Plasmodium yoelli infected tumor-bearing mice, several MDSC- and Treg- recruiting cytokines and growth factors were downregulated in the tumor microenvironment. Plasmodium infection could inhibit tumor secretion of Granulocyte-Macrophage Colony-Stimulating Factor human (GMCSF), IL-10, IL-6, C-C class chemokines (CCL)-17, and CCL-22 through the release of exosome-like vesicles [94]. The CCL17/22-C-C chemokine receptor4 (CCR4) pathway is the major pathway in the recruitment of Tregs into the tumor microenvironment [94]. Besides, attenuated T. gondii infections do not inhibit the recruitment of Treg cells. Foxp3 expressing cells do not change in the Pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment [95]. Moreover, non-replicating Toxoplasma uracil auxotrophs (NATUA) and Carbomyl phosphate synthetase (cps) gene knockout strains show that these parasites can enhance the T cell infiltration and improve the tumor immunogenicity of the tumor. For instance, combination therapy of attenuated T. gondii NRTUA with anti-programmed death-1 (PD-1) led to elevation of CD8+ T cell infiltration mediated by dendritic cell-secreted IL-12 and to tumor-specific IFN-γ production in the PDAC tumor microenvironment [96]. CD8+ T cell infiltration in the TME will be reduced with the depletion of IL-12. Combination therapy of NRTUA with anti-PD-1 also significantly reduced tumor weight in PDAC solid tumor [96].

8. Conclusions and Future Directions

In this review, we have gathered information about antitumor effects induced by protozoan parasites, and the associations between infection with protozoan parasites and cancers are well-evidenced. Parasites and some derived products may exert beneficial antitumor responses which could serve as new strategies to treat and prevent these diseases. Perhaps these antineoplastic effects induced by the protozoan parasite reflect the evolution to protect itself as well as the integrity of the host. As mentioned in Table 1, these parasites, including T gondii, Plasmodium, Leishmania, N. caninum, T. cruzi, and Eimeria, and also the secreting proteins, have direct or indirect effects on cancer cells or on the tumor microenvironment, resulting in halting cancer growth or eliminating cancer in mice models. However, the mechanisms involved in cancer development regulation triggered by protozoans are diverse and not yet fully described, and even the related types of tumors are very limited. Even though protozoan parasites are unicellular eukaryotes with similar sharps, the host cell, life cycle, and virulence are distinct, thus the activated immune effects on carcinogenesis are not the same. Apart from the above factors, the variability of antitumor responses may count on the following factors: the type of cancer cell and its transformation stage or even the tissue type and location in which it is found, the inoculation method, the vitality of parasites, as well as the regulated immune responses generated by the host. Most protozoan parasites could elicit unspecific activation of immune cells, i.e., DC and NK through different signaling pathways, and induce antitumor cytokines, such as IFN-γ, IL-12, and TNF-α.
Nevertheless, these antitumor studies are mainly limited to animal models, there is no data evidence from clinical trials to support these results in human patients. The consideration of such parasite-dependent intervention focuses on the harm to the host and unexpected infection caused by injecting live parasites, which may hamper the results of clinical trials, and lead to a practical problem about how to avoid immune damage caused by parasitic infection when using the infection to fight tumors. Of note is the famous example that T. cruzi and its extracts which exhibited toxic effects over different tumors both in experimental animals and humans about 80 years ago. However, the complicated political and international situation interrupted this work. One possible solution to this concern is to identify parasite-derived products and associated molecules with toxic effects on cancer cells and on the microenvironment or that possess binding cancer cell features. Several in vitro and in vivo studies demonstrated that a malaria protein rVAR2 can directly bind with distinct ofCS from a wide range of different cancer cell lines, as confirmed by intravenous injection of rVAR2 in xenograft animal models. Furthermore, the calreticulin of T. cruzi can directly bind with endothelial cells and work as a potent angiogenesis inhibitor through a VEGF-dependent pathway. It is an impractical idea, at least now, to replace drug treatment with derived parasitic products given the fact that only limited clinical evidence was obtained based on these molecules, but the features of these products open the possibilities via acting as adjuvants or drug carriers to conduct comprehensive or combination therapies for improving the effectiveness of current drugs on cancers. In line with this idea, according to the preliminary in vitro analysis, a hemiasterlin analog (KT886) conjugated rVAR2 (VDC886) is demonstrated to effectively kill a total of 33 cancer cells, and in addition, the efficacy of VDC886 treatment is confirmed as effective by inhibiting the growth of karpas299 and PC-3 cancer in mouse models. Unlike the live parasite, the antitumor effect of VDC886 is mainly dependent on the drug KT886, as the cancers were effectively bound by rVAR2 in a concentration and CS-dependent manner rather than the involved signaling of HLA molecules. Therefore, it is essential to identify more parasitic derivative products and search for more effective drugs for potential use in antitumor treatments, as well as to illustrate their actions in regulating immune signaling.
Another major concern is the effectiveness of parasitic immunotherapy. To what level of targeting ability can clinical parasitic infection reach? Up to now, most antitumor studies of live attenuated protozoan parasites were focused on Plasmodium and Toxoplasma strains. As for T. gondii, crucial genes of the de novo pyrimidine synthesis pathways were genetically disrupted or knocked out so that the attenuated strains can normally invade host cells but lose the intracellular proliferation ability, thus eliminating the potential harm to normal host cells. In accordance with that mentioned above, the contingent risk or side effect can be outweighed compared with the fatal results of most malignant tumors out of the fact that these protozoan parasites are opportunistic pathogens, let alone the applied strains being genetically or gamma radiation-attenuated with limited pathogenicity. In addition, the immunotherapeutic attenuated T. gondii strains have been demonstrated in several mouse models by peritoneal or intratumoral injection by activating native and adaptive immune responses and affecting the tumor microenvironment. Thus, key research should be conducted for the clinical use of immunotherapeutic attenuated protozoan parasites in the next few years. It is worth noting that three clinical trials of Plasmodium immunotherapy for advanced lung cancer (NCT02786589), breast and liver cancers (NCT03474822), and advanced cancers (NCT03375983) have been approved and are ongoing in China. In recent years, cancer immunotherapy, especially the adaptive chimeric antigen receptor T (CAR-T) cell therapy, and immune checkpoint blockade therapy have been recognized as leading breakthroughs. Imagine if we combined CAR-T and checkpoint blockade therapy with attenuated protozoan parasites? For example, Plasmodium and Toxoplasma strains expressed with particular ‘chimeric antigen receptor’, i.e., PD-1 or CTLA-4, so as to increase the targeting ability of cancer cells. In this way, the ‘cold’ tumors can be turned into ‘hot’ ones by the infection of parasites, and the inactivated ‘sleeping’ immune cells can be wakened by checkpoint inhibitors and able to recognize and attack cancer cells. The so-called CAR-Plasmodium or CAR-Toxoplasma may be an impractical crazy idea or it may be of great potential in clinical use.
In spite of the potential use of protozoan-based antitumor therapy, it is important to mention the occurrence of the side effects in the clinical use of live parasites. For example, the infection of T. gondii [97], T. cruzi [98], or Plasmodium [99] may trigger cachexia which remains a devastating problem for cancer patients. As a complex manifestation of systemic inflammation, gut barrier dysfunction, and muscle wasting, cancer cachexia affects the survival and quality of life of patients [100]. One possible solution is to use attenuated pathogens through gene modification, such as T. gondii so that the pathogens would be cleared before the induction of other side effects when the host developed acquired immunity (around 1–2 weeks). However, the short duration of infection by the attenuated pathogen may be unable to induce sustainable activation of the immune system. Thus, several doses of attenuated parasites are necessary to achieve anticancer efficacy in clinical use. Another solution is to give a single low dose of a drug, such as artesunate for Plasmodium [41], to control the parasite density to a safe level. This may raise the concerns of the drug resistance of artesunate. According to their reports [41], up to now there are no signs of drug resistance in the treatment of over 100 cancer patients, therefore the clinical safety of Plasmodium immunotherapy is guaranteed.
Table 1. Protozoan parasites with antitumor effect.
Table 1. Protozoan parasites with antitumor effect.
ParasiteCancerMechanism of ActionReference
Leishmania spp.breast cancer; HPV-associated tumorsActivation of CD4+ and CD8+ T cells, macrophages in spleen or NK cell; Induction of proinflammatory cytokines that help the generation of protective Th1 responses; Increasing the levels of IgG2a[6,81]
Neospora caninummurine thymoma EG7Activation of NK cell- and CD8-T cell-dependent protective antitumor response; IFN-γ secretion in tumor microenvironment[7]
Eimeria spp.Sarcoma tumor S180EA upregulates inflammatory modulators MCP-1, IL-6, IL-12, IFN-γ, and TNF-α[26]
Toxoplasma gondiiMelanoma; Lewis lung carcinoma; Ehrlich’s adenocarcinoma; Pancreatic ductal adenocarcinoma; ovarian carcinomaSecreted proteins activate antitumor immune responses involving CD4+ and CD8+ T cells, IL-12, IFN-γ and TNF-α or activation of NK cell; Increase the levels of IgG1 and IgG2a; Suppressed the levels of angiogenic factors (VEGF, integrin, MMP2, and MMP9)[8,12,23,60,95]
Trypanosoma cruzimammary cancer; colon cancer; Melanoma; Ehrlich’s adenocarcinomaCalreticulin inhibits vascular endothelial growth factor (VEGF)-induced cell proliferation and induces cell apoptosis; activation of CD4+ and CD8+ T cells and macrophages and DC[30,101,102,103]
Plasmodium spp.non-Hodgkin’s lymphoma (Karpas299) and prostate cancer (PC-3); Lewis lung cancer; hepatocellular carcinoma; breast cancerrVAR2 binds with the distinct oncofetal chondroitin sulfate that makes rVAR2 a potential ideal carrier for anti-cancer drug delivery; Activation of NK cell, DC, CD8+ T cell; Suppressed the levels of angiogenic factors (VEGF, MMP9, IGF); Reduce the numbers of MDSC and Treg through CCL17/22-CCR4 pathway[18,104,105,106]
Above all, the use of genetically modified protozoan parasites and derived molecules combined with chemotherapy could be an important element for promoting antitumor treatment.

Author Contributions

Conceptualization, S.L. and X.Z.; data curation, writing—original draft preparation, H.D., S.W. and Z.J.; writing—review and editing, B.Z., Y.H. and K.H.; visualization and supervision, S.W. and X.Z.; visualization and validation, H.D. and Z.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Natural Science Foundation of China (No. 81802037) the Natural Science Foundation of Zhejiang Province (No. LY22H190003), the Zhejiang Province Medical and Health Science and Technology Plan (No. 2022KY138 and 2022RC125), specific scientific research funds of Hangzhou Medical College (Zhejiang Academy of Medical Sciences) (No. YS2021008), Basic Scientific Research Funds of Department of Education of Zhejiang Province (No. KYZD202104 and KYYB202101).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Schirrmacher, V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). Int. J. Oncol. 2019, 54, 407–419. [Google Scholar] [CrossRef] [PubMed]
  3. Roskin, G. Toxin therapy of experimental cancer; the influence of protozoan infections upon transplanted cancer. Cancer Res. 1946, 6, 363–365. [Google Scholar] [PubMed]
  4. Hauschka, T.S.; Goodwin, M.B. Trypanosoma cruzi Endotoxin (KR) in the Treatment of Malignant Mouse Tumors. Science 1948, 107, 600–602. [Google Scholar] [CrossRef]
  5. Ramirez-Toloza, G.; Abello, P.; Ferreira, A. Is the Antitumor Property of Trypanosoma cruzi Infection Mediated by Its Calreticulin? Front. Immunol. 2016, 7, 268. [Google Scholar] [CrossRef]
  6. Caner, A.; Sadiqova, A.; Erdogan, A.; Namlises, D.; Nalbantsoy, A.; Oltulu, F.; Toz, S.; Yigitturk, G.; Ozkok, E.; Gunduz, C.; et al. Targeting of antitumor immune responses with live-attenuated Leishmania strains in breast cancer model. Breast Cancer 2020, 27, 1082–1095. [Google Scholar] [CrossRef]
  7. Lantier, L.; Poupée-Beaugé, A.; Di Tommaso, A.; Ducournau, C.; Epardaud, M.; Lakhrif, Z.; Germon, S.; Debierre-Grockiego, F.; Mévélec, M.-N.; Battistoni, A.; et al. Neospora caninum: A new class of biopharmaceuticals in the therapeutic arsenal against cancer. J. Immunother. Cancer 2020, 8, e001242. [Google Scholar] [CrossRef]
  8. Zhu, Y.C.; Elsheikha, H.M.; Wang, J.H.; Fang, S.; He, J.J.; Zhu, X.Q.; Chen, J. Synergy between Toxoplasma gondii type I DeltaGRA17 immunotherapy and PD-L1 checkpoint inhibition triggers the regression of targeted and distal tumors. J. Immunother. Cancer 2021, 9, e002970. [Google Scholar] [CrossRef]
  9. Bhattacharya, P.; Dey, R.; Dagur, P.K.; Kruhlak, M.; Ismail, N.; Debrabant, A.; Joshi, A.B.; Akue, A.; Kukuruga, M.; Takeda, K.; et al. Genetically Modified Live Attenuated Leishmania donovani Parasites Induce Innate Immunity through Classical Activation of Macrophages That Direct the Th1 Response in Mice. Infect. Immun. 2015, 83, 3800–3815. [Google Scholar] [CrossRef]
  10. Kochanowsky, J.A.; Koshy, A.A. Toxoplasma gondii. Curr. Biol. 2018, 28, R770–R771. [Google Scholar] [CrossRef]
  11. Suzuki, Y.; Kobayashi, A. Antitumor effect of intralesional injection with formalin-fixed toxoplasma gondii organisms on lewis lung carcinoma in toxoplasma-infected mice. Cancer Lett. 1985, 25, 247–254. [Google Scholar] [CrossRef]
  12. Hafez, E.N.; Moawed, F.S.M.; Abdel-Hamid, G.R.; Elbakary, N.M. Gamma Radiation-Attenuated Toxoplasma gondii Provokes Apoptosis in Ehrlich Ascites Carcinoma-Bearing Mice Generating Long-Lasting Immunity. Technol. Cancer Res. Treat. 2020, 19, 1533033820926593. [Google Scholar] [CrossRef]
  13. Li, Y.; Zhang, Y.; Xia, N.; Zhou, T.; Shen, B. Antitumor effects of a Toxoplasma mutant lacking lactate dehydrogenases. Parasitol. Res. 2021, 120, 3335–3339. [Google Scholar] [CrossRef]
  14. Baird, J.R.; Fox, B.A.; Sanders, K.L.; Lizotte, P.H.; Cubillos-Ruiz, J.R.; Scarlett, U.K.; Rutkowski, M.R.; Conejo-Garcia, J.R.; Fiering, S.; Bzik, D.J. Avirulent Toxoplasma gondii Generates Therapeutic Antitumor Immunity by Reversing Immunosuppression in the Ovarian Cancer Microenvironment. Cancer Res. 2013, 73, 3842–3851. [Google Scholar] [CrossRef]
  15. Sugahara, K.; Mikami, T.; Uyama, T.; Mizuguchi, S.; Nomura, K.; Kitagawa, H. Recent advances in the structural biology of chondroitin sulfate and dermatan sulfate. Curr. Opin. Struct. Biol. 2003, 13, 612–620. [Google Scholar] [CrossRef]
  16. Shao, C.; Shi, X.; White, M.; Huang, Y.; Hartshorn, K.; Zaia, J. Comparative glycomics of leukocyte glycosaminoglycans. FEBS J. 2013, 280, 2447–2461. [Google Scholar] [CrossRef]
  17. Clausen, T.M.; Pereira, M.A.; Al Nakouzi, N.; Oo, H.Z.; Agerbæk, M.Ø.; Lee, S.; Ørum-Madsen, M.S.; Kristensen, A.R.; El-Naggar, A.; Grandgenett, P.M.; et al. Oncofetal Chondroitin Sulfate Glycosaminoglycans Are Key Players in Integrin Signaling and Tumor Cell Motility. Mol. Cancer Res. 2016, 14, 1288–1299. [Google Scholar] [CrossRef]
  18. Salanti, A.; Clausen, T.M.; Agerbæk, M.Ø.; Al Nakouzi, N.; Dahlbäck, M.; Oo, H.Z.; Lee, S.; Gustavsson, T.; Rich, J.R.; Hedberg, B.J.; et al. Targeting Human Cancer by a Glycosaminoglycan Binding Malaria Protein. Cancer Cell 2015, 28, 500–514. [Google Scholar] [CrossRef]
  19. Clausen, T.M.; Christoffersen, S.; Dahlbäck, M.; Langkilde, A.E.; Jensen, K.E.; Resende, M.; Agerbæk, M.; Andersen, D.; Berisha, B.; Ditlev, S.B.; et al. Structural and Functional Insight into How the Plasmodium falciparum VAR2CSA Protein Mediates Binding to Chondroitin Sulfate A in Placental Malaria. J. Biol. Chem. 2012, 287, 23332–23345. [Google Scholar] [CrossRef]
  20. Agerbaek, M.O.; Bang-Christensen, S.R.; Yang, M.H.; Clausen, T.M.; Pereira, M.A.; Sharma, S.; Ditlev, S.B.; Nielsen, M.A.; Choudhary, S.; Gustavsson, T.; et al. The VAR2CSA malaria protein efficiently retrieves circulating tumor cells in an EpCAM-independent manner. Nat. Commun. 2018, 9, 3279. [Google Scholar] [CrossRef]
  21. Dlugonska, H. Toxoplasma Rhoptries: Unique Secretory Organelles and Source of Promising Vaccine Proteins for Immunoprevention of Toxoplasmosis. J. Biomed. Biotechnol. 2008, 2008, 632424. [Google Scholar] [CrossRef]
  22. Panas, M.W.; Boothroyd, J.C. Seizing control: How dense granule effector proteins enable Toxoplasma to take charge. Mol. Microbiol. 2021, 115, 466–477. [Google Scholar] [CrossRef]
  23. Fox, B.A.; Sanders, K.L.; Rommereim, L.M.; Guevara, R.B.; Bzik, D.J. Secretion of Rhoptry and Dense Granule Effector Proteins by Nonreplicating Toxoplasma gondii Uracil Auxotrophs Controls the Development of Antitumor Immunity. PLoS Genet. 2016, 12, e1006189. [Google Scholar] [CrossRef]
  24. Wang, P.; Li, S.; Zhao, Y.; Zhang, B.; Li, Y.; Liu, S.; Du, H.; Cao, L.; Ou, M.; Ye, X.; et al. The GRA15 protein from Toxoplasma gondii enhances host defense responses by activating the interferon stimulator STING. J. Biol. Chem. 2019, 294, 16494–16508. [Google Scholar] [CrossRef]
  25. Seo, S.H.; Kim, S.G.; Shin, J.H.; Ham, D.W.; Shin, E.H. Toxoplasma GRA16 Inhibits NF-kappaB Activation through PP2A-B55 Upregulation in Non-Small-Cell Lung Carcinoma Cells. Int. J. Mol. Sci. 2020, 21, 6642. [Google Scholar] [CrossRef]
  26. Rosenberg, B.; Juckett, D.A.; Aylsworth, C.F.; Dimitrov, N.V.; Ho, S.-C.; Judge, J.W.; Kessel, S.; Quensen, J.; Wong, K.-P.H.; Zlatkin, I.; et al. Protein from intestinal Eimeria protozoan stimulates IL-12 release from dendritic cells, exhibits antitumor properties in vivo and is correlated with low intestinal tumorigenicity. Int. J. Cancer 2004, 114, 756–765. [Google Scholar] [CrossRef]
  27. Junqueira, C.; Santos, L.I.; Galvão-Filho, B.; Teixeira, S.M.; Rodrigues, F.G.; DaRocha, W.D.; Chiari, E.; Jungbluth, A.A.; Ritter, G.; Gnjatic, S.; et al. Trypanosoma cruzi as an effective cancer antigen delivery vector. Proc. Natl. Acad. Sci. USA 2011, 108, 19695–19700. [Google Scholar] [CrossRef]
  28. Ramírez, G.; Valck, C.; Aguilar, L.; Kemmerling, U.; López-Muñoz, R.; Cabrera, G.; Morello, A.; Ferreira, J.; Maya, J.D.; Galanti, N.; et al. Roles of Trypanosoma cruzi calreticulin in parasite–host interactions and in tumor growth. Mol. Immunol. 2012, 52, 133–140. [Google Scholar] [CrossRef]
  29. Molina, M.C.; Ferreira, V.; Valck, C.; Aguilar, L.; Orellana, J.; Rojas, A.; Ramirez, G.; Billetta, R.; Schwaeble, W.; Lemus, D.; et al. An in vivo role for Trypanosoma cruzi calreticulin in antiangiogenesis. Mol. Biochem. Parasitol. 2005, 140, 133–140. [Google Scholar] [CrossRef]
  30. López, N.C.; Valck, C.; Ramírez, G.; Rodríguez, M.; Ribeiro, C.; Orellana, J.; Maldonado, I.; Albini, A.; Anacona, D.; Lemus, D.; et al. Antiangiogenic and Antitumor Effects of Trypanosoma cruzi Calreticulin. PLoS Neglected Trop. Dis. 2010, 4, e730. [Google Scholar] [CrossRef]
  31. Shah, M.A.K.A.S.A.A.A.; Kamal, M.A.; Akhtar, S. Tumor Angiogenesis and VEGFR-2: Mechanism, Pathways and Current Biological Therapeutic Interventions. Curr. Drug Metab. 2021, 22, 50–59. [Google Scholar] [CrossRef] [PubMed]
  32. Al-Ostoot, F.H.; Salah, S.; Khamees, H.A.; Khanum, S.A. Tumor Angiogenesis: Current Challenges and Therapeutic Opportunities (Review). Cancer Treat. Res. Commun. 2021, 28, 100422. [Google Scholar] [CrossRef] [PubMed]
  33. Shu, Q.; Li, W.; Li, H.; Sun, G. Vasostatin Inhibits VEGF-Induced Endothelial Cell Proliferation, Tube Formation and Induces Cell Apoptosis under Oxygen Deprivation. Int. J. Mol. Sci. 2014, 15, 6019–6030. [Google Scholar] [CrossRef] [PubMed]
  34. Gajewski, T.F.; Schreiber, H.; Fu, Y.-X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef]
  35. Gabrilovich, D.; Pisarev, V. Tumor escape from immune response: Mechanisms and targets of activity. Curr. Drug Targets 2003, 4, 525–536. [Google Scholar] [CrossRef]
  36. Chiossone, L.; Dumas, P.-Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688, Correction in 2018, 18, 726. [Google Scholar] [CrossRef]
  37. Hammer, Q.; Rückert, T.; Borst, E.M.; Dunst, J.; Haubner, A.; Durek, P.; Heinrich, F.; Gasparoni, G.; Babic, M.; Tomic, A.; et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018, 19, 453–463. [Google Scholar] [CrossRef]
  38. Sivori, S.; Pende, D.; Quatrini, L.; Pietra, G.; Della Chiesa, M.; Vacca, P.; Tumino, N.; Moretta, F.; Mingari, M.C.; Locatelli, F.; et al. NK cells and ILCs in tumor immunotherapy. Mol. Asp. Med. 2020, 80, 100870. [Google Scholar] [CrossRef]
  39. Chen, L.; He, Z.; Qin, L.; Li, Q.; Shi, X.; Zhao, S.; Chen, L.; Zhong, N.; Chen, X. Antitumor Effect of Malaria Parasite Infection in a Murine Lewis Lung Cancer Model through Induction of Innate and Adaptive Immunity. PLoS ONE 2011, 6, e24407. [Google Scholar] [CrossRef]
  40. Korbel, D.S.; Newman, K.C.; Almeida, C.R.; Davis, D.M.; Riley, E.M. Heterogeneous Human NK Cell Responses to Plasmodium falciparum-Infected Erythrocytes. J. Immunol. 2005, 175, 7466–7473. [Google Scholar] [CrossRef]
  41. Chen, X.; Qin, L.; Hu, W.; Adah, D. The mechanisms of action of Plasmodium infection against cancer. Cell Commun. Signal. 2021, 19, 74. [Google Scholar] [CrossRef] [PubMed]
  42. Sanabria, M.X.H.; Vargas-Inchaustegui, D.A.; Xin, L.; Soong, L. Role of Natural Killer Cells in Modulating Dendritic Cell Responses to Leishmania amazonensis Infection. Infect. Immun. 2008, 76, 5100–5109. [Google Scholar] [CrossRef] [PubMed]
  43. Baird, J.R.; Byrne, K.T.; Lizotte, P.H.; Toraya-Brown, S.; Scarlett, U.K.; Alexander, M.P.; Sheen, M.R.; Fox, B.A.; Bzik, D.J.; Bosenberg, M.; et al. Immune-Mediated Regression of Established B16F10 Melanoma by Intratumoral Injection of Attenuated Toxoplasma gondii Protects against Rechallenge. J. Immunol. 2012, 190, 469–478. [Google Scholar] [CrossRef] [PubMed]
  44. Park, E.; Patel, S.; Wang, Q.; Andhey, P.; Zaitsev, K.; Porter, S.; Hershey, M.; Bern, M.; Plougastel-Douglas, B.; Collins, P.; et al. Toxoplasma gondii infection drives conversion of NK cells into ILC1-like cells. ELife 2019, 8, e47605. [Google Scholar] [CrossRef]
  45. Moro, K.; Yamada, T.; Tanabe, M.; Takeuchi, T.; Ikawa, T.; Kawamoto, H.; Furusawam, J.; Ohtani, M.; Fujii, H.; Koyasu, S. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature 2010, 463, 540–544. [Google Scholar] [CrossRef]
  46. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef]
  47. Kamiya, T.; Chang, Y.-H.; Campana, D. Expanded and Activated Natural Killer Cells for Immunotherapy of Hepatocellular Carcinoma. Cancer Immunol. Res. 2016, 4, 574–581. [Google Scholar] [CrossRef]
  48. Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef]
  49. Liu, E.; Tong, Y.; Dotti, G.; Shaim, H.; Savoldo, B.; Mukherjee, M.; Orange, J.; Wan, X.; Lu, X.; Reynolds, A.; et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2017, 32, 520–531. [Google Scholar] [CrossRef]
  50. Takeuchi, Y.; Nishikawa, H. Roles of regulatory T cells in cancer immunity. Int. Immunol. 2016, 28, 401–409. [Google Scholar] [CrossRef]
  51. Choi, Y.; Shi, Y.; Haymaker, C.L.; Naing, A.; Ciliberto, G.; Hajjar, J. T-cell agonists in cancer immunotherapy. J. Immunother. Cancer 2020, 8, e000966. [Google Scholar] [CrossRef]
  52. Smith-Garvin, J.E.; Koretzky, G.A.; Jordan, M.S. T cell activation. Annu. Rev. Immunol. 2009, 27, 591–619. [Google Scholar] [CrossRef]
  53. Okada, M.; Shimizu, K.; Fujii, S.-I. Identification of Neoantigens in Cancer Cells as Targets for Immunotherapy. Int. J. Mol. Sci. 2022, 23, 2594. [Google Scholar] [CrossRef]
  54. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef]
  55. Neefjes, J.; Jongsma, M.L.M.; Paul, P.; Bakke, O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat. Rev. Immunol. 2011, 11, 823–836. [Google Scholar] [CrossRef]
  56. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  57. Baitsch, L.; Baumgaertner, P.; Devêvre, E.; Raghav, S.K.; Legat, A.; Barba, L.; Wieckowski, S.; Bouzourene, H.; Deplancke, B.; Romero, P.; et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Investig. 2011, 121, 2350–2360. [Google Scholar] [CrossRef]
  58. Dey, R.; Dagur, P.K.; Selvapandiyan, A.; McCoy, J.P.; Salotra, P.; Duncan, R.; Nakhasi, H.L. Live attenuated Leishmania donovani p27 gene knockout parasites are nonpathogenic and elicit long-term protective immunity in BALB/c mice. J. Immunol. 2013, 190, 2138–2149. [Google Scholar] [CrossRef]
  59. Dey, R.; Natarajan, G.; Bhattacharya, P.; Cummings, H.; Dagur, P.K.; Terrazas, C.; Selvapandiyan, A.; McCoy, J.P.; Duncan, R.; Satoskar, A.R.; et al. Characterization of Cross-Protection by Genetically Modified Live-Attenuated Leishmania donovani Parasites against Leishmania mexicana. J. Immunol. 2014, 193, 3513–3527. [Google Scholar] [CrossRef]
  60. Sanders, K.L.; Fox, B.A.; Bzik, D.J. Attenuated Toxoplasma gondii therapy of disseminated pancreatic cancer generates long-lasting immunity to pancreatic cancer. Oncoimmunology 2016, 5, e1104447. [Google Scholar] [CrossRef]
  61. Verneau, J.; Sautés-Fridman, C.; Sun, C.-M. Dendritic cells in the tumor microenvironment: Prognostic and theranostic impact. Semin. Immunol. 2020, 48, 101410. [Google Scholar] [CrossRef]
  62. Mildner, A.; Jung, S. Development and Function of Dendritic Cell Subsets. Immunity 2014, 40, 642–656. [Google Scholar] [CrossRef]
  63. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef]
  64. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
  65. De Veer, M.J.; Curtis, J.M.; Baldwin, T.M.; DiDonato, J.A.; Sexton, A.; McConville, M.J.; Handman, E.; Schofield, L. MyD88 is essential for clearance ofLeishmania major: Possible role for lipophosphoglycan and Toll-like receptor 2 signaling. Eur. J. Immunol. 2003, 33, 2822–2831. [Google Scholar] [CrossRef]
  66. Campos, M.A.S.; Almeida, I.C.; Takeuchi, O.; Akira, S.; Valente, E.P.; Procópio, D.O.; Travassos, L.R.; Smith, J.A.; Golenbock, D.T.; Gazzinelli, R.T. Activation of Toll-Like Receptor-2 by Glycosylphosphatidylinositol Anchors from a Protozoan Parasite. J. Immunol. 2001, 167, 416–423. [Google Scholar] [CrossRef]
  67. Krishnegowda, G.; Hajjar, A.M.; Zhu, J.; Douglass, E.J.; Uematsu, S.; Akira, S.; Woods, A.S.; Gowda, D.C. Induction of proinflammatory responses in macrophages by the glycosylphosphatidylinositols of Plasmodium falciparum: Cell signaling receptors, glycosylphosphatidylinositol (GPI) structural requirement, and regulation of GPI activity. J. Biol. Chem. 2005, 280, 8606–8616. [Google Scholar] [CrossRef]
  68. Debierre-Grockiego, F.; Azzouz, N.; Schmidt, J.; Dubremetz, J.F.; Geyer, H.; Geyer, R.; Weingart, R.; Schmidt, R.R.; Schwarz, R.T. Roles of glycosylphosphatidylinositols of Toxoplasma gondii. Induction of tumor necrosis factor-alpha production in macrophages. J. Biol. Chem. 2003, 278, 32987–32993. [Google Scholar] [CrossRef]
  69. Yarovinsky, F.; Zhang, D.; Andersen, J.F.; Bannenberg, G.L.; Serhan, C.N.; Hayden, M.S.; Hieny, S.; Sutterwala, F.S.; Flavell, R.A.; Ghosh, S.; et al. TLR11 Activation of Dendritic Cells by a Protozoan Profilin-Like Protein. Science 2005, 308, 1626–1629. [Google Scholar] [CrossRef]
  70. Conlon, K.C.; Miljkovic, M.D.; Waldmann, T.A. Cytokines in the Treatment of Cancer. J. Interferon Cytokine Res. 2019, 39, 6–21. [Google Scholar] [CrossRef]
  71. Jorgovanovic, D.; Song, M.; Wang, L.; Zhang, Y. Roles of IFN-gamma in tumor progression and regression: A review. Biomark Res. 2020, 8, 49. [Google Scholar] [CrossRef] [PubMed]
  72. Lu, X. Impact of IL-12 in Cancer. Curr. Cancer Drug Targets 2017, 17, 682–697. [Google Scholar] [CrossRef] [PubMed]
  73. Batchu, R.B.; Gruzdyn, O.V.; Kolli, B.K.; Dachepalli, R.; Umar, P.S.; Rai, S.K.; Singh, N.; Tavva, P.S.; Weaver, D.W.; Gruber, S.A. IL-10 Signaling in the Tumor Microenvironment of Ovarian Cancer. Adv. Exp. Med. Biol. 2021, 1290, 51–65. [Google Scholar] [CrossRef] [PubMed]
  74. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, Y.; Liu, L.; Zhou, Y.; Zhou, P.; Yan, Q.; Chen, X.; Ding, S.; Zhu, F. CKLF1 Enhances Inflammation-Mediated Carcinogenesis and Prevents Doxorubicin-Induced Apoptosis via IL6/STAT3 Signaling in HCC. Clin. Cancer Res. 2019, 25, 4141–4154. [Google Scholar] [CrossRef] [PubMed]
  76. Li, L.; Yu, R.; Cai, T.; Chen, Z.; Lan, M.; Zou, T.; Wang, B.; Wang, Q.; Zhao, Y.; Cai, Y. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int. Immunopharmacol. 2020, 88, 106939. [Google Scholar] [CrossRef] [PubMed]
  77. Kim, J.-O.; Jung, S.-S.; Kim, S.-Y.; Kim, T.Y.; Shin, D.-W.; Lee, J.-H.; Lee, Y.-H. Inhibition of Lewis Lung Carcinoma Growth by Toxoplasma gondii through Induction of Th1 Immune Responses and Inhibition of Angiogenesis. J. Korean Med Sci. 2007, 22, S38–S46. [Google Scholar] [CrossRef] [PubMed]
  78. Hafez, E.N.; Youssef, H.; El-Kabany, H.A. Vaccination with gamma radiation-attenuated Toxoplasma gondii protects against ovarian infiltration in mice-bearing Ehrlich ascites carcinoma. Int. J. Radiat. Biol. 2020, 96, 814–822. [Google Scholar] [CrossRef]
  79. Astaneh, M.; Dashti, S.; Esfahani, Z.T. Humoral immune responses against cancer-testis antigens in human malignancies. Hum. Antibodies 2019, 27, 237–240. [Google Scholar] [CrossRef]
  80. Rudnick, S.I.; Lou, J.; Shaller, C.C.; Tang, Y.; Klein-Szanto, A.J.; Weiner, L.M.; Marks, J.D.; Adams, G.P. Influence of Affinity and Antigen Internalization on the Uptake and Penetration of Anti-HER2 Antibodies in Solid Tumors. Cancer Res. 2011, 71, 2250–2259. [Google Scholar] [CrossRef]
  81. Salehi, M.; Taheri, T.; Mohit, E.; Zahedifard, F.; Seyed, N.; Taslimi, Y.; Sattari, M.; Bolhassani, A.; Rafati, S. Recombinant Leishmania tarentolae encoding the HPV type 16 E7 gene in tumor mice model. Immunotherapy 2012, 4, 1107–1120. [Google Scholar] [CrossRef]
  82. Zenina, A.V.; Kravtsov, E.G.; Tsetsegsaikhan, B.; Yashina, N.V.; Dalin, M.V.; Karpenko, L.P.; Sheklakova, L.A.; Kallinikova, V.D. The study of immunological component in antitumor effect of Trypanosoma cruzi. Bull. Exp. Biol. Med. 2008, 145, 352–354. [Google Scholar] [CrossRef]
  83. Tas, S.W.; Maracle, C.; Balogh, E.; Szekanecz, Z. Targeting of proangiogenic signalling pathways in chronic inflammation. Nat. Rev. Rheumatol. 2015, 12, 111–122. [Google Scholar] [CrossRef]
  84. Wang, B.; Li, Q.; Wang, J.; Zhao, S.; Nashun, B.; Qin, L.; Chen, X. Plasmodium infection inhibits tumor angiogenesis through effects on tumor-associated macrophages in a murine implanted hepatoma model. Cell Commun. Signal. 2020, 18, 157. [Google Scholar] [CrossRef]
  85. Shieh, Y.-S.; Hung, Y.-J.; Hsieh, C.-B.; Chen, J.-S.; Chou, K.-C.; Liu, S.-Y. Tumor-Associated Macrophage Correlated with Angiogenesis and Progression of Mucoepidermoid Carcinoma of Salivary Glands. Ann. Surg. Oncol. 2008, 16, 751–760. [Google Scholar] [CrossRef]
  86. Ferreira, V.; Valck, C.; Sánchez, G.; Gingras, A.; Tzima, S.; Molina, M.C.; Sim, R.; Schwaeble, W.; Ferreira, A. The Classical Activation Pathway of the Human Complement System Is Specifically Inhibited by Calreticulin from Trypanosoma cruzi. J. Immunol. 2004, 172, 3042–3050. [Google Scholar] [CrossRef]
  87. Ramírez, G.; Valck, C.; Molina, M.C.; Ribeiro, C.H.; López, N.; Sánchez, G.; Ferreira, V.P.; Billetta, R.; Aguilar, L.; Maldonado, I.; et al. Trypanosoma cruzi calreticulin: A novel virulence factor that binds complement C1 on the parasite surface and promotes infectivity. Immunobiology 2011, 216, 265–273. [Google Scholar] [CrossRef]
  88. Pyo, K.-H.; Jung, B.-K.; Chai, J.-Y.; Shin, E.-H. Suppressed CD31 Expression in Sarcoma-180 Tumors after Injection with Toxoplasma gondii Lysate Antigen in BALB/c Mice. Korean J. Parasitol. 2010, 48, 171–174. [Google Scholar] [CrossRef]
  89. Vonderheide, R.H.; Bayne, L.J. Inflammatory networks and immune surveillance of pancreatic carcinoma. Curr. Opin. Immunol. 2013, 25, 200–205. [Google Scholar] [CrossRef]
  90. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  91. Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2016, 27, 109–118. [Google Scholar] [CrossRef]
  92. Zhang, Y.; Velez-Delgado, A.; Mathew, E.; Li, D.; Mendez, F.M.; Flannagan, K.; Rhim, A.D.; Simeone, D.M.; Beatty, G.L.; Pasca di Magliano, M. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017, 66, 124–136. [Google Scholar] [CrossRef]
  93. Wang, X.; Lang, M.; Zhao, T.; Feng, X.; Zheng, C.; Huang, C.; Hao, J.; Dong, J.; Luo, L.; Li, X.; et al. Cancer-FOXP3 directly activated CCL5 to recruit FOXP3+Treg cells in pancreatic ductal adenocarcinoma. Oncogene 2016, 36, 3048–3058. [Google Scholar] [CrossRef]
  94. Adah, D.; Yang, Y.; Liu, Q.; Gadidasu, K.; Tao, Z.; Yu, S.; Dai, L.; Li, X.; Zhao, S.; Qin, L.; et al. Plasmodium infection inhibits the expansion and activation of MDSCs and Tregs in the tumor microenvironment in a murine Lewis lung cancer model. Cell Commun. Signal. 2019, 17, 32. [Google Scholar] [CrossRef]
  95. Payne, S.N.; Emmerich, P.B.; Davis, N.M.; Deming, D.A.; Knoll, L.J. Novel Murine Pancreatic Tumor Model Demonstrates Immunotherapeutic Control of Tumor Progression by a Toxoplasma gondii Protein. Infect. Immun. 2021, 89, e00508-21. [Google Scholar] [CrossRef]
  96. Bahwal, S.A.; Chen, J.J.; Lilin, E.; Hao, T.; Chen, J.; Carruthers, V.B.; Lai, J.; Zhou, X. Attenuated Toxoplasma gondii enhances the antitumor efficacy of anti-PD1 antibody by altering the tumor microenvironment in a pancreatic cancer mouse model. J. Cancer Res. Clin. Oncol. 2022; online ahead of print. [Google Scholar] [CrossRef]
  97. Hatter, J.A.; Kouche, Y.M.; Melchor, S.J.; Ng, K.; Bouley, D.M.; Boothroyd, J.C.; Ewald, S.E. Toxoplasma gondii infection triggers chronic cachexia and sustained commensal dysbiosis in mice. PLoS ONE 2018, 13, e0204895. [Google Scholar] [CrossRef]
  98. Truyens, C.; Torrico, F.; Angelo-Barrios, A.; Lucas, R.; Heremans, H.; De Baetselier, P.; Carlier, Y. The cachexia associated with Trypanosoma cruzi acute infection in mice is attenuated by anti-TNF-alpha, but not by anti-IL-6 or anti-IFN-gamma antibodies. Parasite Immunol. 1995, 17, 561–568. [Google Scholar] [CrossRef]
  99. Lamb, T.; Brown, D.E.; Potocnik, A.J.; Langhorne, J. Insights into the immunopathogenesis of malaria using mouse models. Expert Rev. Mol. Med. 2006, 8, 1–22. [Google Scholar] [CrossRef]
  100. Herremans, K.M.; Riner, A.N.; Cameron, M.E.; Trevino, J.G. The Microbiota and Cancer Cachexia. Int. J. Mol. Sci. 2019, 20, 6267. [Google Scholar] [CrossRef]
  101. Ubillos, L.; Freire, T.; Berriel, E.; Chiribao, M.L.; Chiale, C.; Festari, M.F.; Medeiros, A.; Mazal, D.; Rondan, M.; Bollati-Fogolin, M.; et al. Trypanosoma cruzi extracts elicit protective immune response against chemically induced colon and mammary cancers. Int. J. Cancer 2016, 138, 1719–1731. [Google Scholar] [CrossRef] [PubMed]
  102. Guzmán, L.A.A.; Lobos-Gonzalez, L.; Rosas, C.; Vallejos, G.; Falcón, C.; Sosoniuk, E.; Coddou, F.; Leyton, L.; Lemus, D.; Quest, A.G.; et al. Human Survivin and Trypanosoma cruzi Calreticulin Act in Synergy against a Murine Melanoma In Vivo. PLoS ONE 2014, 9, e95457. [Google Scholar] [CrossRef]
  103. Atayde, V.D.; Jasiulionis, M.G.; Cortez, M.; Yoshida, N. A recombinant protein based on Trypanosoma cruzi surface molecule gp82 induces apoptotic cell death in melanoma cells. Melanoma Res. 2008, 18, 172–183. [Google Scholar] [CrossRef] [PubMed]
  104. Pan, J.; Ma, M.; Qin, L.; Kang, Z.; Adah, D.; Tao, Z.; Li, X.; Dai, L.; Zhao, S.; Chen, X.; et al. Plasmodium infection inhibits triple negative 4T1 breast cancer potentially through induction of CD8+ T cell-mediated antitumor responses in mice. Biomed. Pharmacother. 2021, 138, 111406. [Google Scholar] [CrossRef]
  105. Liang, Y.; Chen, X.; Tao, Z.; Ma, M.; Adah, D.; Li, X.; Dai, L.; Ding, W.; Fanuel, S.; Zhao, S.; et al. Plasmodium infection prevents recurrence and metastasis of hepatocellular carcinoma possibly via inhibition of the epithelial-mesenchymal transition. Mol. Med. Rep. 2021, 23, 418. [Google Scholar] [CrossRef]
  106. Zhou, D.; Zheng, H.; Liu, Q.; Lu, X.; Deng, X.; Jiang, L.; Hou, B.; Fu, Y.; Zhu, F.; Ding, Y.; et al. Attenuated plasmodium sporozoite expressing MAGE-A3 induces antigen-specific CD8+ T cell response against lung cancer in mice. Cancer Biol. Med. 2019, 16, 288–298. [Google Scholar]
Figure 1. The landscape of the host’s immune system activated by parasites against the tumor. There are three ways that parasites could stimulate the host’s immune system to eliminate tumors. (1) Plasmodium could boost the level of CD69/25 and promote the natural killer T cell activation to secrete TNF-α; (2) RVAR2-KT886, a targeted drug based Plasmodium rVAR2, could recognize the CS in the surface of tumor cells and then kill it; (3) Toxoplasma gondii, Leishmania, Trypanosoma cruzi, Neospora caninum and Plasmodium could boost the level of CD80/86 and promote the dendritic cell maturity, and the mature dendritic cell actives CD4+ T cell to be Help1 T cell or Help2 T cell through MHC II. Help1 T cell would kill tumor through IL-12/IFN-gamma Axis; Help2 T cell would stimulate the B cell to produce specific IgG against tumor cell. Abbreviations: TNF-α: tumor necrosis factor alpha; IFN-γ: Interferon Gamma; IL-12: Interleukin 12; MHC I: major histocompatibility complex class I; MHC II: major histocompatibility complex class II; CS: chondroitin sulfate.
Figure 1. The landscape of the host’s immune system activated by parasites against the tumor. There are three ways that parasites could stimulate the host’s immune system to eliminate tumors. (1) Plasmodium could boost the level of CD69/25 and promote the natural killer T cell activation to secrete TNF-α; (2) RVAR2-KT886, a targeted drug based Plasmodium rVAR2, could recognize the CS in the surface of tumor cells and then kill it; (3) Toxoplasma gondii, Leishmania, Trypanosoma cruzi, Neospora caninum and Plasmodium could boost the level of CD80/86 and promote the dendritic cell maturity, and the mature dendritic cell actives CD4+ T cell to be Help1 T cell or Help2 T cell through MHC II. Help1 T cell would kill tumor through IL-12/IFN-gamma Axis; Help2 T cell would stimulate the B cell to produce specific IgG against tumor cell. Abbreviations: TNF-α: tumor necrosis factor alpha; IFN-γ: Interferon Gamma; IL-12: Interleukin 12; MHC I: major histocompatibility complex class I; MHC II: major histocompatibility complex class II; CS: chondroitin sulfate.
Bioengineering 09 00395 g001
Figure 2. Protozoans modulate the tumor immune microenvironment and inhibit angiogenesis. Leishmania, Neospora caninum and Toxoplasma gondii could downregulate some cytokines (GMCSF, CCL17/22, IL-10 or IL-6) to inhibit Treg and MDSC; besides, these protozoans also could inhibit angiogenesis through suppressing the level of angiogenic factor (VEGF, MMP2/9 or integrin). Among these, the scavenger-like receptor could recognize TcCRT secreted by Trypanosoma cruzi, and it could inhibit VEGF to induce cell apoptosis. Abbreviations: MDSC: myeloid-derived immunosuppressive cell; Treg: regulatory T cells; GMCSF: Granulocyte-Macrophage Colony-Stimulating Factor human; CCL17/22: C-C class chemokines 17/22; IL-10: Interleukin 10; IL-6: Interleukin 6; VEGF: vascular endothelial growth factor; MMP2/9: matrix metallopeptidase 2/9; TcCRT: Trypanosoma cruzi calreticulin.
Figure 2. Protozoans modulate the tumor immune microenvironment and inhibit angiogenesis. Leishmania, Neospora caninum and Toxoplasma gondii could downregulate some cytokines (GMCSF, CCL17/22, IL-10 or IL-6) to inhibit Treg and MDSC; besides, these protozoans also could inhibit angiogenesis through suppressing the level of angiogenic factor (VEGF, MMP2/9 or integrin). Among these, the scavenger-like receptor could recognize TcCRT secreted by Trypanosoma cruzi, and it could inhibit VEGF to induce cell apoptosis. Abbreviations: MDSC: myeloid-derived immunosuppressive cell; Treg: regulatory T cells; GMCSF: Granulocyte-Macrophage Colony-Stimulating Factor human; CCL17/22: C-C class chemokines 17/22; IL-10: Interleukin 10; IL-6: Interleukin 6; VEGF: vascular endothelial growth factor; MMP2/9: matrix metallopeptidase 2/9; TcCRT: Trypanosoma cruzi calreticulin.
Bioengineering 09 00395 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ding, H.; Wu, S.; Jin, Z.; Zheng, B.; Hu, Y.; He, K.; Lu, S.; Zhuo, X. Anti-Tumor Effect of Parasitic Protozoans. Bioengineering 2022, 9, 395. https://doi.org/10.3390/bioengineering9080395

AMA Style

Ding H, Wu S, Jin Z, Zheng B, Hu Y, He K, Lu S, Zhuo X. Anti-Tumor Effect of Parasitic Protozoans. Bioengineering. 2022; 9(8):395. https://doi.org/10.3390/bioengineering9080395

Chicago/Turabian Style

Ding, Haojie, Songrui Wu, Zi Jin, Bin Zheng, Yuan Hu, Ke He, Shaohong Lu, and Xunhui Zhuo. 2022. "Anti-Tumor Effect of Parasitic Protozoans" Bioengineering 9, no. 8: 395. https://doi.org/10.3390/bioengineering9080395

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop