Skip to main content

Perspective view of allogeneic IgG tumor immunotherapy

Abstract

Allogeneic tumors are eradicated by host immunity; however, it is unknown how it is initiated until the report in Nature by Yaron Carmi et al. in 2015. Currently, we know that allogeneic tumors are eradicated by allogeneic IgG via dendritic cells. AlloIgG combined with the dendritic cell stimuli tumor necrosis factor alpha and CD40L induced tumor eradication via the reported and our proposed potential signaling pathways. AlloIgG triggers systematic immune responses targeting multiple antigens, which is proposed to overcome current immunotherapy limitations. The promising perspectives of alloIgG immunotherapy would have advanced from mouse models to clinical trials; however, there are only 6 published articles thus far. Therefore, we hope this perspective view will provide an initiative to promote future discussion.

Introduction

Immunotherapy has achieved durable responses in some cancer patients. However, the tumor microenvironment poses significant challenges that limit its effectiveness by creating an immunosuppressive milieu that shields tumors from antitumor immunity, thereby preventing many patients from benefiting from these therapies. Therefore, there is an urgent need to find new therapies to improve patient outcomes. As we reported in the Journal of Hematology and Oncology in 2020, dendritic cells (DCs) play a significant role in initiating and maintaining the immune response against cancer cells [1]. These specialized cells can recognize and process antigens, present them to T cells, and regulate immune responses upon uptake of immune complexes (ICs), making them attractive targets for cancer immunotherapy.

The Fc region of the antibody contains constant sequences, with only a small number of variants, and can elicit a host of cellular responses by binding to various Fc receptors expressed widely by different leukocytes. Fc–FcγR interactions represent a key component of the in vivo activity of therapeutic mAbs [2, 3]. The interaction of IgG–FcγR activates various downstream immune regulatory pathways with multiple functional consequences, including activation of DCs and T cells [4]. Carmi et al. found that allogeneic IgG (alloIgG) can combine with dendritic cells to induce a powerful T-cell antitumor response [5]. This review and perspective review introduces the effects and safety of alloIgG tumor immunotherapy and the underlying mechanisms for its potential future clinical application.

Immunoglobulin G and FcγR are important in immunity

Immunoglobulin G (IgG) comprises 10–20% of all plasma proteins and 70–75% of total immunoglobulins [6]. Its high antigen affinity, somatic hypermutation, and essential role in immune memory are well established. IgG includes IgG1, IgG2, IgG3, and IgG4 subtypes. IgG1 and IgG3 have the highest affinity for type I Fcγ receptors (FcγRs) for increased cytotoxic activity in vivo, while IgG2 and IgG4 have poor affinity with all type I FcγRs [7]. In addition to activating C1q, IgG has multiple functions, including binding to FcγRs on immune cells.

When multimeric IgG immune complexes interact with activated FcγRs, receptor clustering and aggregation occur, leading to phosphorylation of the ITAM structural domain by SRC family kinases (such as LYN, LCK, HCK, and FGR) and the recruitment and activation of SYK family kinases [8, 9]. This event activates the PI3K-PKC pathway, resulting in Ca2+ mobilization and cellular activation [10, 11]. MEK and MAP family kinases and the Ras pathway are then activated [12]. The IgG–FcγR interaction and downstream signaling may lead to antibody-dependent cellular cytotoxicity (ADCC) or phagocytosis (ADCP), cytokine and chemokine release, leukocyte differentiation and survival and T/B-cell responses [13,14,15].

Furthermore, IgG can directly neutralize toxins and microbes [16]. IgG can also generate inflammatory mediators and eliminate opsonized microbes [17]. An IgG molecule contains two variable Fab domains for antigen binding, one constant Fc domain for FcγR binding and a hinge region in a Y shape [18]. Although the Fc domain has conventionally been considered the invariant domain of an IgG molecule, it exhibits remarkable structural heterogeneity with different IgG subclasses and biantennary N-linked glycans [19]. These structural determinants modulate the conformational flexibility of the IgG Fc domain and impact its ability to bind to different types of FcγRs (type I or type II) [20].

FcγRs are widely expressed on immune cells and specifically bind to the IgG Fc domain [21]. Activating FcγRs include FcγRI, FcγRIIA, FcγRIIIA, and FcγRIIIB (CD64, CD32a, CD16a, CD16b) in humans and FcγRI, FcγRIII, and FcγRIV in mice [7]. A single inhibitory receptor, FcγRIIB (also known as CD32b), is activated by a tyrosine inhibition motif (ITIM) in both humans and mice [22, 23]. Activating FcγR mRNA is expressed in monocytes, macrophages, and monocyte-derived DCs (moDCs), and inhibitory FcγRIIB mRNA is expressed in mouse cDCs, plasmacytoid DCs (pDCs), moDCs and macrophages. Human cDCs and pDCs express FcγRIIB mRNA as well as FcγRIIA. Both mouse and human CD172α+ cDCs express low levels of FcγRI, as determined by flow cytometry [13, 20]. Although mRNA expression does not always predict protein expression, recent human and mouse flow cytometry data support these findings [24]. These data suggest that macrophages and moDCs express mRNA for most of the activating and inhibitory FcγRs, whereas cDCs and pDCs primarily express mRNA for the inhibitory FcγRIIB.

The relative expression of activating and inhibiting FcγRs coexpressed on many immune cells determines the activation threshold of immune cell responses [25]. FcγRIIb activation leads to receptor cross-linking, phosphorylation by SRC family kinases and phosphatase recruitment to their ITIM structural domains [26, 27]. ITIM-recruited phosphatases (SHIP1 and SHP2) lead to the hydrolysis of phosphatidylinositol 3,4,5-triphosphate (PIP3) to phosphatidylinositol 4,5-biphosphate (PIP2), inhibiting PLCγ and the tyrosine kinase BTK [27,28,29].

IgG mAbs tumor immunotherapy requires IgG Fc–DC FcγR interaction

Therapeutic monoclonal antibodies (mAbs) interact with innate and adaptive immunity in vivo [7]. Therapeutic mAbs bind to cancer cell surface antigens, inhibiting their proliferation and survival [30]. The IgG Fc–FcγR interaction mediates ADCC, ADCP, and CDC functions to block growth signals and angiogenesis and activate the immune response [7, 31]. Despite the diverse mechanisms of action of therapeutic mAbs, a common function is their interaction with FcγRs expressed on the surface of leukocytes through their Fc domain. FcγRIIIa significantly improves the therapeutic efficacy of anti-CD20 monoclonal antibodies [32]. B-cell lymphoma, breast cancer, and colorectal cancer patients carrying FcγRIIa and FcγRIIIa allelic variants are more responsive to antitumor antibody therapy [33,34,35,36]. In HER2+ breast cancer, mAbs have become the frontline standard of care, outperforming HER2-specific small molecule inhibitors and achieving excellent responses with modest toxicities [37], which require IgG–FcγR interaction [38]. Anti-GITR antibodies were found to require activating FcγRs [39]. Fc–FcγR interactions can promote innate immunity via cellular differentiation and survival. Fc–FcγR interactions promote antigen processing and presentation and the maturation and activation of dendritic cells [4]. Last, B cells are also regulated by Fc–FcγR interactions by their type I FcγR, FcγRIIb and type II FcγR, CD23 [40]. These results suggest that Fc–FcγR interactions are vital in cancer immunotherapy.

IgG Fc–DC FCγR interaction activates DCs and T cells

DCs are the most effective antigen-presenting cells (APCs) [41]. DCs include type 1 cDCs (cDC1s), type 2 cDCs (cDC2s), and pDCs. DCs become activated upon exposure to foreign antigens, which can occur through the engagement of conserved bacterial or viral antigens known as pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs) [42]. Resting immature DCs (imDCs) express PRRs of Toll-like receptors (TLRs), membrane-associated C-type lectin receptors, and mannose receptors [43,44,45]. DC maturation is regulated by activating and inhibitory type I FcγRs. Steady-state DCs express both inhibitory FcγRIIb and activating FcγRIIa, which prevents inappropriate or uncontrolled DC maturation [7]. Selective blockade of FcγRIIB using monoclonal antibodies leads to human DC maturation [46]. imDCs become mature, losing their endocytic capacity but increasing their antigen processing and presentation capacity [47]. maDCs upregulate chemokine receptors such as CCR7, driving their homing to lymph nodes [48], where they present antigens to naive CD4+ or CD8+ T cells (Fig. 1a) [49, 50]. cDC1s cross-present antigens to cytotoxic CD8+ T cells and promote the activation of CD4+ T helper type 1 (Th1) cells, while cDC2s induce CD4+ T-cell responses [51]. pDCs in the blood and spleen express MHC class II and costimulatory molecules [52]. Newly identified moDCs are present in mouse and human tumors [53]. MoDCs in tissues have a limited capacity to transport antigens to lymph nodes and activate naive T cells in vitro, which distinguishes them from Ly6C+ or CD14hi monocytes [54, 55]. Therefore, it is unclear to what extent moDCs contribute to the initiation of new T-cell responses.

Fig. 1
figure 1

IC–FCγR interactions lead to DC antigen presentation and T-cell activation. a When exposed to foreign antigens, PRRS and FcγRs can mediate the induction of dendritic cell maturation. During maturation, imDCs lose their endocytic capacity while increasing their capacity for antigen processing and presentation, driving their homing to lymph nodes, where they present antigens to naive CD4+ or CD8+ T cells. b ICs bind to FcγRs on the surface of DCs, are internalized and processed, and subsequently bind to neonatal FcRn, trafficking and MHC-mediated antigen presentation. c Matching of TCR with MHC molecules on DCs. d The activation of a T-cell depends on its interaction with APCs and requires three signals

Activated FcγRs promote degradative antigen processing and presentation, thereby activating T cells, while internalization by FcγRIIB tends to preserve the intact antigen for subsequent transfer to B cells. ICs are bound to FcγRs on the surface of DCs, internalized and subsequently bound to the neonatal Fc receptor (FcRn). Since FcRn is predominantly intracellular and binds IgG at acidic pH, it is well placed to engage IgG–ICs within endolysosomal compartments and regulate IgG–IC trafficking and MHC-mediated antigen presentation (Fig. 1b) [56]. ICs are more effective in antigen-presenting functions of DCs than free antigens [56, 57]. In mouse studies, DCs from splenic mice showed more efficient uptake of ovalbumin (OVA) preincubated with anti-ovalbumin IgG than “naked” OVA. Notably, OVA:IgG immune complexes induced CD4+ and CD8+ T-cell proliferation more effectively than “naked” OVA in mice transplanted with OVA-specific CD8+ or CD4+ T cells [58]. Inactivated Francisella tularensis immune complexes (mAb-iFt) are a more protective vaccine against lethal tularemia than iFt alone. Nelson et al. discovered that targeting iFt to FcγRs via mAb-iFt leads to enhanced DC maturation, with FcγR being needed for mAb-iFt-induced maturation of bone marrow-derived DCs [59]. Fc–FcγR interactions hold promise for DC-specific vaccination-based strategies [7, 60].

The activation of a T-cell depends on its interaction with APCs and requires three signals (Fig. 1b–d). Signal 1 is the specific peptide recognized by the T-cell receptor (TCR). Major histocompatibility complex (MHC) molecules (either MHC-I for cytotoxic CD8+ T cells or MHC-II for CD4+ T cells) are needed (Fig. 1c) [61]. The intracellular pathways [1, 50] mediate antigen degradation and peptide loading onto MHC molecules. Antigen presentation to CD4+ T cells is enhanced only when the antigen and IgG are present within the same phagosome [24]. Signal 2 is the costimulatory molecules on T cells (e.g., CD28) and their receptors on APCs, such as CD80 and CD86 (B7.1 and B7.2). Signal 3 is the cytokines needed to define the type of response. These three signals induce Ag-specific CD4+ or CD8+ T-cell responses (Fig. 1d) [62]. DCs can ingest virus-infected or tumor cells and present Ags to specific CD8+ T cells via cross-presentation through an MHC-I pathway [63].

Tumors are eradicated by allogeneic IgG via DCs

The combination of tumor-binding alloIgG and DCs has been shown to effectively eradicate both primary and metastatic mouse tumors, including melanoma, pancreatic, lung, colon, and breast cancer (Table 1) [5]. In syngeneic C57BL/6 mice, B16 melanoma cells proliferated, while they were rejected in allogeneic 129S1 mice, with all animals treated by other methods experiencing rapid tumor recurrence. Allogeneic transplanted tumors had more mature myeloid DCs that were more activated than syngeneic tumors. IgM and IgG antibodies binding to allogeneic tumors enabled tumor-infiltrating DCs to process and present tumor antigens to CD4+ T cells, and this response was abrogated in FcγR-deficient mice. Only allogeneic immunoglobulin-IC could activate bone marrow-derived DCs (BMDCs) in vitro, and BMDC activated by alloIgG-IC induced significant T cell proliferation [5]. However, only minor effects were observed when alloIgG was injected into tumors in autologous mice in vivo. The possible explanation of the limited effect could be the difference between BMDCs and tumor-associated dendritic cells (TADCs) as described below.

Table 1 AlloIgG tumor immunotherapy

AlloIgG combined with DC stimuli TNFα and CD40L eradicated tumors

Unlike BMDCs, TADCs did not respond to alloIgG against tumor cells or lysate (alloIgG-IC) (Fig. 2a, b). However, PolyI:C, TNFα+CD40L, or IFNγ+CD40L could activate TADCs to take up and present alloIgG-IC. Intratumoral injection of alloIgG combined with TNFα+CD40L or PolyI:C eliminated B16 and LL/2 homologous tumors. In mouse models, alloIgG+CD40+TNFα almost eliminated melanoma and breast cancer metastases (Fig. 2c). Culturing malignant pleural mesothelioma patients BMDCs with allogeneic IgG activated and enhanced autologous CD4+ T-cell propagation. Finally, the authors found that alloIgG isolated from healthy donors could similarly induce TADC activation in the presence of tumor necrosis factor alpha (TNF-α) and CD40 when cultured with tumor cells, validating the clinical performance of this approach. These results suggest that TADC unresponsiveness to IC is not due to the suppressive nature of the tumor microenvironment but rather a consequence of normal monocyte maturation [5]. Analysis of the signaling pathways in MoDC, TADCs, and BMDCs indicates that rapid Syk phosphorylation following ligation of FcγRs with ICs induces dramatic downstream protein activation in the MAPK (p38, pJNK, pERK) and PI3K/Akt (pAkt) pathways in BMDCs (Fig. 3a) [64]. Although SHP-1 regulates DC activation and Syk phosphorylation, inhibition of SHP-1 alone is not sufficient to induce MoDC or TADC to respond to IC. But it requires both PTEN and SHP-1/SHIP-1 for MoDCs and TADCs to activation by alloIgG-ICs (Fig. 3b) [64].

Fig. 2
figure 2

AlloIgG combined with TNFα and CD40L induced complete elimination of tumor cells. a Syngeneic BMDCs loaded with AlloIgG-IC activate T cells and prevent tumor recurrence in mice. b When AlloIgG was injected into tumors in autologous mice, TADC cannot transmit signals through their Fcγ receptor after contact with AlloIgG-IC in a highly immunosuppressed tumor microenvironment. c Combining tumor-binding AlloIgG with TNFα and CD40L enables TADC to internalize tumor antigens via the Fcγ receptor. These antigens are then processed by DCs and presented to T cells, which attack primary tumors and distant metastases

Fig. 3
figure 3

Mechanisms of DC-mediated signaling by alloIgG action. a Stimulation of BMDCs with AlloIgG-IC resulted in a significant increase in phosphorylated MAPK p38, ERK1/2, and JNK, as well as robust phosphorylation of Akt. b Once monocytes are released from their bone marrow niche into the circulation, they markedly elevate the levels of phosphorylated SHP-1 and phosphatases that regulate Akt activation. c Simultaneous blockade of SHP-1 and Akt-regulating phosphatases (such as PTEN and SHIP-1) enables activation of TADCs and MoDCs. Green arrows represent our proposed potential TADC/MoDC activation signaling pathways by TNFα and CD40L

Hypothetical potential signaling pathways in alloIgG-CD40-TNF-α-activated DCs

Next, we hypothetically propose the signaling pathways in alloIgG-CD40-TNF-α-activated DCs for the first time as a perspective view for further discussion (Fig. 3). We believe the mechanistic study will provide a basis for its future improvement. CD40, as a tumor necrosis factor, primes DCs for effective and specific T-cell activation [65]. Activation of DCs with CD40 agonists increased survival and cytokine secretion of IL-1, IL-6, IL-8, IL-12, TNF-α, and macrophage inflammatory protein-1α and upregulated costimulatory molecules of MHC class II, LFA-3, CD80, and CD86, promoting antigen presentation, priming, and cross-priming of T helper cells and cytotoxic T lymphocytes, respectively [66]. Based on their study, Vidalain and colleagues propose a model of CD40-mediated signaling in human DCs that includes CD40-induced membrane raft reorganization and the recruitment of TNFR-associated factors 2 and 3 (TRAFs) and activation of Lyn and other Src family kinases. Lyn activation leads to IL-1α, IL-1β, and IL-1Ra mRNA expression through a MEK/ERK pathway. Activation of p38 MAPK, which induces the expression of IL-12 mRNA, is likely stimulated through a TRAF-initiated pathway and, to some extent, through a Src family kinase-dependent pathway in the early phase of CD40 signaling [67]. Additionally, TRAF activates the NF-κB, MAPK, PI3K, and PLCγ pathways [68].

The essential role of CD40L in the induction of protective tumor immunity led researchers to expect that agonistic anti-CD40 antibodies would act as potent adjuvants to promote tumor immunity. CD40-stimulated DCs significantly induce T-cell proliferation and cytokine production [69]. CD40 engagement provides survival signals to DCs, making them resistant to Fas ligand expressed by activated T cells [70, 71]. CD40 agonistic antibodies generated CTL responses that eradicated lymphoma tumors. CD40 ligation could overcome peptide-induced peripheral CTL tolerance and increase antitumor efficacy [72,73,74]. Evidence suggests that the effect of anti-CD40 antibodies on CD40-expressing cells critically depends on whether they interact with FcγR and C1 [75]. Therefore, the CD40/CD40L interaction is necessary for optimal antigen presentation by DCs. However, some studies suggest the opposite result, indicating that CD40/CD40L may be closely associated with tumorigenesis [76]. CD40 is expressed on the surface of normal cells and cancer cells of the bladder, lung, and ovary [77,78,79] and is highly expressed in malignant hematological tumors [80]. CD40L is highly expressed in many cancers, but its tumorigenic functions in neoplastic disease remain controversial [81].

TNF-α is a potent anticancer cytokine that binds to two receptors, TNFRSF1A (TNFR1) in all cell types and TNFRSF1B (TNFR2) in immune cells. Activation of the NF-κB, JNK, p38 MAPK, ERK, and PI3K pathways by TNF-α binding to TNFR2 guides cell proliferation and survival [82] (Fig. 3c). Despite its multiple functions, TNFα can have conflicting effects on cancer cells. As demonstrated by Carswell, elevated levels of TNFα can eliminate MCA-induced sarcomas, and approximately 28% of cancers are sensitive to sTNFα [83]. Direct intratumor injection of DCs into homologous mouse tumors can reverse established tumor nodules in mice and provide effective immunity against subsequent tumor threats. This antitumor effect can be enhanced by prepriming DCs with recombinant TNF-α [84]. Cancer cell secretion of TNFα can promote DC production, differentiation, and maturation [85]. However, low levels of TNFα expression may be protumorigenic, as reviewed in detail by Balkwill [86].

AlloIgG triggers systematic immune responses targeting multiple antigens

It has been found that alloIgG binding specifically to tumor cells, rather than the source of IgG or their cross-linking with syngeneic IgG, induces strong immune responses [5]. Syngeneic IgG bound only six B16 membrane proteins, but alloIgG preferentially bound 16 cell membrane proteins, including transmembrane glycoprotein NMB (GP-NMB) [5]. GP-NMB antibodies, αCD40 and TNFα together activate DCs and induce FcγR-dependent tumor regression with activated effector/memory T-cell infiltration, suggesting that tumor-reactive T cells targeting tumor-associated antigens that are not widely expressed alloantigens are needed [5].

It was shown that alloIgG triggers systematic immune responses [5]. Systematic immune responses were also reported in tumor-specific McAb IgG therapy. Spitzer, Matthew et al. developed an intuitive model, a computational method called scaffold maps. Scaffold map analysis revealed that treatment of a spontaneous model of carcinom MMTV-PyMT triple-negative breast cancer with anti-PD-1 antibodies triggered only a transient immune response at the local tumor, but the combination of tumor-binding antibodies and adjuvants triggered both local and system-wide immune responses in this model, including lymph nodes, bone marrow and blood [87]. This could explain why the triple-negative breast cancer model is refractory to checkpoint blockade therapy, whereas the combination of alloIgG-IC with IFNγ and CD40 therapy is effective [87]. Binbin et al. developed a multimodal recurrent neural network called MARIA, which predicts the likelihood of antigen presentation for genes of interest in the context of specific HLA class II alleles. We propose that MARIA may be used to identify candidate antigens more accurately from patient sequencing data to improve alloIgG immunotherapy [88].

AlloIgG immunotherapy may overcome current immunotherapy limitations

Immunotherapy has made considerable progress, with some patients achieving long-lasting responses through IgG McAbs and cellular immunotherapies. Therapeutic antibodies containing Fc domains promote antitumor activity by activating DCs [89]. Fc–FcγR interactions and uptake of ICs by DCs play a vital role in the in vivo activity of APCs and T cells through various mechanisms (Fig. 1) [90]. However, IgG McAb therapy often leads to drug resistance and tumor recurrence in 6 months via many different mechanisms [3]. We propose that alloIgG immunotherapy may overcome the limitations of IgG McAb therapy because it triggers robust systematic immune responses targeting multiple antigens via different signaling pathways (Figs. 2 and 3).

DC immunotherapy and vaccines have gained a crucial position due to their unique ability to present MHC class I and II molecule-restricted peptides and activate T cells (Fig. 1) [1, 91]. DCs are usually from the bone marrow or spleen and are rare. DC vaccines are feasible because DCs can now be cultured in large numbers ex vivo by controlling DC maturation and homing to lymph nodes [92]. A detailed protocol for isolating MoDCs from blood and tumors and activating MoDCs with tumor ICs is available [93]. There are two common methods to clinically prepare DC vaccines: loading tumor antigens directly onto DC cells or fusing DCs with tumor cells [94]. However, to date, DC-based vaccines have not achieved the expected therapeutic efficacy [95]. We propose that alloIgG immunotherapy or antigen-alloIgG complex-stimulated DC vaccines may cause the missing piece of the DC vaccine immunotherapy puzzle.

The efficacy and safety perspectives of alloIgG immunotherapy

One limitation of alloIgG immunotherapy is that it works effectively only in tumors smaller than 20 mm2 in mouse models and becomes almost inert once the tumor exceeds an average size of approximately 40 mm2 due to tumor-infiltrating MoDC apoptosis [96]. One potential reason for this resistance might be the formation of cell-in-cell structures in large tumors [97]. Gutwillig et al. investigated the combination of dendritic cell adjuvants and tumor binding anti-TRP1 antibodies to treat a mouse model of relapse and found that the tumor cells remaining after immunotherapy form unique cell-in-cell structures and generate a membrane architecture that is impenetrable by immune-derived lytic granules, cytotoxic compounds, and chemotherapies. While reactive T cells can often kill the outer cells in this structure, the inner cells remain viable and intact, surviving for weeks in culture containing these T cells. Once the T cells are removed, the inner tumor cells disseminate back, suggesting that this biological process may be a central mechanism through which tumor cells evade T-cell immunity and give rise to relapsed tumors [97]. To increase the antitumor effects of alloIgG, we propose that alloIgG may be used in combination with other therapies, such as chemotherapy, radiotherapy, immune checkpoint inhibitors and CD4+ T cells. Several studies have shown that combining McAb IgG with conventional chemotherapy and radiotherapy can improve efficacy. For example, stereotactic body radiotherapy enhances the antitumor effects of the anti-PD-L1 McAb durvalumab in patients with early-stage NSCLC [98], and the combination of McAb pembrolizumab and radiotherapy has shown promising activity in patients with triple-negative breast cancer [99]. In patients with recurrent nasopharyngeal carcinoma, McAb toripalimab combined with intensity-modulated radiotherapy showed tolerability and promising antitumor activity [100]. Rasoulouniriana et al. discovered that CD4+ T cells isolated from tumors and tumor-binding antibodies have a strong synergistic effect to mediate tumor regression [101].

One of the safety concerns of alloIgG immunotherapy is whether it causes graft-versus-host disease (GVHD) due to genetic variation or polymorphisms among individual persons. AlloIgG immunotherapy exhibits therapeutic efficacy and safety in mouse models, although its promising therapeutic efficacy and safety in humans need to be tested [5, 64, 87, 96, 97, 101, 102]. Its prospective safety in humans may be further suggested by the safe use of allogeneic CAR-T cells, allogeneic γδT cells and natural killer cells [102,103,104,105,106,107,108,109]. Allogeneic γδT cells from haploidentical donors have been utilized to treat hematological malignancies, resulting in complete remission without signs of GVHD [105]. Furthermore, allogeneic Vγ9Vδ2 T-cell immunotherapy has demonstrated clinical safety and extended survival in patients with late-stage lung or liver cancer [107]. One advantage of alloIgG and allogeneic cell immunotherapy is that it allows for the preparation and storage of alloIgG and allogeneic cells in advance, thus reducing the waiting time and cost for patients to receive treatment [109].

Review and view

The IgG Fc–DC FcγR interaction enables antigen recognition, processing and presentation by DCs, which activates T-cell immunity. Allogeneic tumors are eradicated by allogeneic IgG via DCs. AlloIgG combined with DC stimuli TNFα and CD40L induced tumor eradication via the reported and prospective signaling pathways. AlloIgG triggers systematic immune responses targeting multiple antigens, which was proposed to overcome current immunotherapy limitations. The promising efficacy and safety perspectives of alloIgG immunotherapy need to be validated.

With more efforts and breakthroughs, we believe that alloIgG tumor immunotherapy has promising potential to demonstrate efficiency and safety in mouse models, enter clinical trials and benefit tumor patients in the future. It has been 8 years since the first report of the important alloIgG tumor immunotherapy in Nature by Stanford University; however, there are only 6 directly related articles published mainly in mouse models (Table 1) [5, 64, 87, 96, 97, 101]. Therefore, we hope this perspective view of alloIgG tumor immunotherapy will provide an initiative to promote future discussion.

Availability of data and materials

Not applicable.

Abbreviations

DCs:

Dendritic cells

ICs:

Immune complexes

alloIgG:

Allogeneic IgG

IgG:

Immunoglobulin G

FcγRs:

Fcγ receptors

ADCC:

Antibody-dependent cellular cytotoxicity

ADCP:

Antibody-dependent cellular phagocytosis

moDC:

Monocyte-derived DC

pDCs:

Plasmacytoid DCs

mAbs:

Monoclonal antibodies

APCs:

Antigen-presenting cells

cDC1s:

Type 1 cDCs

cDC2s:

Type 2 cDCs

PAMPs:

Pathogen-associated molecular patterns

PRRs:

Pattern recognition receptors

imDCs:

Immature DCs

TLRs:

Toll-like receptors

FcRn:

Neonatal Fc receptor

OVA:

Ovalbumin

mAb-iFt:

Nactivated Francisella tularensis immune complexes

TCR:

T-cell receptor

MHC:

Major histocompatibility complex

BMDCs:

Bone marrow-derived dcs

TADCs:

Tumor-associated dendritic cells

TNF-α:

Tumor necrosis factor alpha

TRAFs:

TNFR-associated factors

GP-NMB:

Glycoprotein NMB

GVHD:

Graft-versus-host disease

References

  1. Wang Y, et al. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol. 2020;13(1):107.

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  2. Barnhart BC, Quigley M. Role of Fc–FcγR interactions in the antitumor activity of therapeutic antibodies. Immunol Cell Biol. 2017;95(4):340–6.

    Article  CAS  PubMed  Google Scholar 

  3. Cai WQ, et al. The latest battles between EGFR monoclonal antibodies and resistant tumor cells. Front Oncol. 2020;10:1249.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Bournazos S, Ravetch JV. Fcγ receptor pathways during active and passive immunization. Immunol Rev. 2015;268(1):88–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Carmi Y, et al. Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity. Nature. 2015;521(7550):99–104.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014;5:520.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Bournazos S, Ravetch JV. Fcγ receptor function and the design of vaccination strategies. Immunity. 2017;47(2):224–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Durden DL, et al. The Fc gamma RI receptor signals through the activation of hck and MAP kinase. J Immunol. 1995;154(8):4039–47.

    Article  CAS  PubMed  Google Scholar 

  9. Durden DL, Liu YB. Protein-tyrosine kinase p72syk in Fc gamma RI receptor signaling. Blood. 1994;84(7):2102–8.

    Article  CAS  PubMed  Google Scholar 

  10. Kanakaraj P, et al. Phosphatidylinositol-3 kinase activation induced upon Fc gamma RIIIA-ligand interaction. J Exp Med. 1994;179(2):551–8.

    Article  CAS  PubMed  Google Scholar 

  11. Ninomiya N, et al. Involvement of phosphatidylinositol 3-kinase in Fc gamma receptor signaling. J Biol Chem. 1994;269(36):22732–7.

    Article  CAS  PubMed  Google Scholar 

  12. Bracke M, et al. Analysis of signal transduction pathways regulating cytokine-mediated Fc receptor activation on human eosinophils. J Immunol. 1998;161(12):6768–74.

    Article  CAS  PubMed  Google Scholar 

  13. Nimmerjahn F, Ravetch JV. Fcγ receptors as regulators of immune responses. Nat Rev Immunol. 2008;8(1):34–47.

    Article  CAS  PubMed  Google Scholar 

  14. Pincetic A, et al. Type I and type II Fc receptors regulate innate and adaptive immunity. Nat Immunol. 2014;15(8):707–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. de Taeye SW, et al. FcγR binding and ADCC activity of human IgG allotypes. Front Immunol. 2020;11:740.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Noris M, Remuzzi G. Overview of complement activation and regulation. Semin Nephrol. 2013;33(6):479–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bournazos S, DiLillo DJ, Ravetch JV. The role of Fc–FcγR interactions in IgG-mediated microbial neutralization. J Exp Med. 2015;212(9):1361–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nimmerjahn F, Ravetch JV. Fc-receptors as regulators of immunity. Adv Immunol. 2007;96:179–204.

    Article  CAS  PubMed  Google Scholar 

  19. Jefferis R. Glycosylation of recombinant antibody therapeutics. Biotechnol Prog. 2005;21(1):11–6.

    Article  CAS  PubMed  Google Scholar 

  20. Sondermann P, et al. The 3.2-A crystal structure of the human IgG1 Fc fragment-Fc γRIII complex. Nature. 2000;406(6793):267–73.

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat Rev Immunol. 2020;20(10):633–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bajtay Z, et al. Expression and role of Fc- and complement-receptors on human dendritic cells. Immunol Lett. 2006;104(1–2):46–52.

    Article  CAS  PubMed  Google Scholar 

  23. de Jong JM, et al. Murine Fc receptors for IgG are redundant in facilitating presentation of immune complex derived antigen to CD8+ T cells in vivo. Mol Immunol. 2006;43(13):2045–50.

    Article  PubMed  Google Scholar 

  24. Guilliams M, et al. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol. 2014;14(2):94–108.

    Article  CAS  PubMed  Google Scholar 

  25. Bournazos S, et al. Functional and clinical consequences of Fc receptor polymorphic and copy number variants. Clin Exp Immunol. 2009;157(2):244–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Muta T, et al. A 13-amino-acid motif in the cytoplasmic domain of FcγRIIB modulates B-cell receptor signalling. Nature. 1994;368(6466):70–3.

    Article  ADS  CAS  PubMed  Google Scholar 

  27. Pearse RN, et al. SHIP recruitment attenuates FcγRIIB-induced B cell apoptosis. Immunity. 1999;10(6):753–60.

    Article  CAS  PubMed  Google Scholar 

  28. Ono M, et al. Role of the inositol phosphatase SHIP in negative regulation of the immune system by the receptor Fc(γ)RIIB. Nature. 1996;383(6597):263–6.

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Ono M, et al. Deletion of SHIP or SHP-1 reveals two distinct pathways for inhibitory signaling. Cell. 1997;90(2):293–301.

    Article  CAS  PubMed  Google Scholar 

  30. Selvaraj P, et al. Natural killer cell and granulocyte Fc gamma receptor III (CD16) differ in membrane anchor and signal transduction. J Immunol. 1989;143(10):3283–8.

    Article  CAS  PubMed  Google Scholar 

  31. Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer. 2012;12(4):278–87.

    Article  CAS  PubMed  Google Scholar 

  32. Goede V, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370(12):1101–10.

    Article  CAS  PubMed  Google Scholar 

  33. Bibeau F, et al. Impact of FcγRIIa–FcγRIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J Clin Oncol. 2009;27(7):1122–9.

    Article  CAS  PubMed  Google Scholar 

  34. Mellor JD, et al. A critical review of the role of Fc gamma receptor polymorphisms in the response to monoclonal antibodies in cancer. J Hematol Oncol. 2013;6:1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Musolino A, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26(11):1789–96.

    Article  CAS  PubMed  Google Scholar 

  36. Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol. 2003;21(21):3940–7.

    Article  CAS  PubMed  Google Scholar 

  37. Oh DY, Bang YJ. HER2-targeted therapies—a role beyond breast cancer. Nat Rev Clin Oncol. 2020;17(1):33–48.

    Article  CAS  PubMed  Google Scholar 

  38. Musolino A, et al. Role of Fcγ receptors in HER2-targeted breast cancer therapy. J Immunother Cancer. 2022;10(1): e003171.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bulliard Y, et al. Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med. 2013;210(9):1685–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang TT, et al. Anti-HA glycoforms drive B cell affinity selection and determine influenza vaccine efficacy. Cell. 2015;162(1):160–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chen K, et al. Tissue-resident dendritic cells and diseases involving dendritic cell malfunction. Int Immunopharmacol. 2016;34:1–15.

    Article  PubMed  PubMed Central  Google Scholar 

  42. van Vliet SJ, Garcia-Vallejo JJ, van Kooyk Y. Dendritic cells and C-type lectin receptors: coupling innate to adaptive immune responses. Immunol Cell Biol. 2008;86(7):580–7.

    Article  PubMed  Google Scholar 

  43. Edwards AD, et al. Toll-like receptor expression in murine DC subsets: lack of TLR7 expression by CD8α+ DC correlates with unresponsiveness to imidazoquinolines. Eur J Immunol. 2003;33(4):827–33.

    Article  CAS  PubMed  Google Scholar 

  44. Fischetti L, et al. The synergistic effects of combining TLR ligand based adjuvants on the cytokine response are dependent upon p38/JNK signalling. Cytokine. 2017;99:287–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li J, et al. The dendritic cell mannose receptor mediates allergen internalization and maturation involving notch 1 signalling. Clin Exp Immunol. 2010;162(2):251–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dhodapkar KM, et al. Selective blockade of inhibitory Fcγ receptor enables human dendritic cell maturation with IL-12p70 production and immunity to antibody-coated tumor cells. Proc Natl Acad Sci USA. 2005;102(8):2910–5.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Whiteside TL, Odoux C. Dendritic cell biology and cancer therapy. Cancer Immunol Immunother. 2004;53(3):240–8.

    Article  PubMed  Google Scholar 

  48. Rodriguez-Fernandez JL, Criado-Garcia O. The chemokine receptor CCR7 uses distinct signaling modules with biased functionality to regulate dendritic cells. Front Immunol. 2020;11:528.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bottcher JP, Reis e Sousa C. The role of type 1 conventional dendritic cells in cancer immunity. Trends Cancer. 2018;4(11):784–92.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Gardner A, Ruffell B. Dendritic cells and cancer immunity. Trends Immunol. 2016;37(12):855–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shen Z, et al. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J Immunol. 1997;158(6):2723–30.

    Article  CAS  PubMed  Google Scholar 

  52. Villadangos JA, Young L. Antigen-presentation properties of plasmacytoid dendritic cells. Immunity. 2008;29(3):352–61.

    Article  CAS  PubMed  Google Scholar 

  53. Coillard A, Segura E. Antigen presentation by mouse monocyte-derived cells: re-evaluating the concept of monocyte-derived dendritic cells. Mol Immunol. 2021;135:165–9.

    Article  CAS  PubMed  Google Scholar 

  54. Tamoutounour S, et al. Origins and functional specialization of macrophages and of conventional and monocyte-derived dendritic cells in mouse skin. Immunity. 2013;39(5):925–38.

    Article  CAS  PubMed  Google Scholar 

  55. Jakubzick C, et al. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity. 2013;39(3):599–610.

    Article  CAS  PubMed  Google Scholar 

  56. Junker F, Gordon J, Qureshi O. Fc gamma receptors and their role in antigen uptake, presentation, and T cell activation. Front Immunol. 2020;11:1393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Junker F, et al. A simple method for measuring immune complex-mediated, Fc gamma receptor dependent antigen-specific activation of primary human T cells. J Immunol Methods. 2018;454:32–9.

    Article  CAS  PubMed  Google Scholar 

  58. Lehmann CHK, et al. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J Exp Med. 2017;214(5):1509–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Nelson NLJ, et al. Fcγ receptors and toll-like receptor 9 synergize to drive immune complex-induced dendritic cell maturation. Cell Immunol. 2019;345: 103962.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Alcaide EG, Krishnarajah S, Junker F. Dendritic cell tumor vaccination via Fc gamma receptor targeting: lessons learned from pre-clinical and translational studies. Vaccines. 2021;9(4):409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wieczorek M, et al. Major histocompatibility complex (MHC) class I and MHC class II proteins: conformational plasticity in antigen presentation. Front Immunol. 2017;8:292.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Raphael I, et al. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine. 2015;74(1):5–17.

    Article  CAS  PubMed  Google Scholar 

  63. Luckashenak N, et al. Constitutive crosspresentation of tissue antigens by dendritic cells controls CD8+ T cell tolerance in vivo. Immunity. 2008;28(4):521–32.

    Article  CAS  PubMed  Google Scholar 

  64. Carmi Y, et al. Akt and SHP-1 are DC-intrinsic checkpoints for tumor immunity. JCI Insight. 2016;1(18): e89020.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Foy TM, et al. Immune regulation by CD40 and its ligand GP39. Annu Rev Immunol. 1996;14:591–617.

    Article  CAS  PubMed  Google Scholar 

  66. Tong AW, Stone MJ. Prospects for CD40-directed experimental therapy of human cancer. Cancer Gene Ther. 2003;10(1):1–13.

    Article  CAS  PubMed  Google Scholar 

  67. Vidalain PO, et al. CD40 signaling in human dendritic cells is initiated within membrane rafts. EMBO J. 2000;19(13):3304–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bishop GA, et al. TRAF proteins in CD40 signaling. Adv Exp Med Biol. 2007;597:131–51.

    Article  PubMed  Google Scholar 

  69. Cella M, et al. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J Exp Med. 1996;184(2):747–52.

    Article  CAS  PubMed  Google Scholar 

  70. Bjorck P, Banchereau J, Flores-Romo L. CD40 ligation counteracts Fas-induced apoptosis of human dendritic cells. Int Immunol. 1997;9(3):365–72.

    Article  CAS  PubMed  Google Scholar 

  71. Koppi TA, et al. CD40 ligand inhibits Fas/CD95-mediated apoptosis of human blood-derived dendritic cells. Eur J Immunol. 1997;27(12):3161–5.

    Article  CAS  PubMed  Google Scholar 

  72. French RR, et al. CD40 antibody evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help. Nat Med. 1999;5(5):548–53.

    Article  CAS  PubMed  Google Scholar 

  73. Diehl L, et al. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments anti-tumor vaccine efficacy. Nat Med. 1999;5(7):774–9.

    Article  CAS  PubMed  Google Scholar 

  74. Sotomayor EM, et al. Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40. Nat Med. 1999;5(7):780–7.

    Article  CAS  PubMed  Google Scholar 

  75. Lang I, Zaitseva O, Wajant H. FcγRs and their relevance for the activity of anti-CD40 antibodies. Int J Mol Sci. 2022;23(21):12869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee GH, et al. The role of CD40 expression in dendritic cells in cancer biology; a systematic review. Curr Cancer Drug Targets. 2014;14(7):610–20.

    Article  CAS  PubMed  Google Scholar 

  77. Cooke PW, et al. CD40 expression in bladder cancer. J Pathol. 1999;188(1):38–43.

    Article  CAS  PubMed  Google Scholar 

  78. Xu W, et al. Anti-tumor activity of gene transfer of the membrane-stable CD40L mutant into lung cancer cells. Int J Oncol. 2010;37(4):935–41.

    CAS  PubMed  Google Scholar 

  79. Gallagher NJ, et al. CD40 activation in epithelial ovarian carcinoma cells modulates growth, apoptosis, and cytokine secretion. Mol Pathol. 2002;55(2):110–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Korniluk A, Kemona H, Dymicka-Piekarska V. Multifunctional CD40L: pro- and anti-neoplastic activity. Tumour Biol. 2014;35(10):9447–57.

    Article  CAS  PubMed  Google Scholar 

  81. Hassan GS, Stagg J, Mourad W. Role of CD154 in cancer pathogenesis and immunotherapy. Cancer Treat Rev. 2015;41(5):431–40.

    Article  CAS  PubMed  Google Scholar 

  82. Sughra K, et al. Interaction of the TNFR-receptor associated factor TRAF1 with I-kappa B kinase-2 and TRAF2 indicates a regulatory function for NF-kappa B signaling. PLoS ONE. 2010;5(9): e12683.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  83. Greish K, Taurin S, Morsy MA. The effect of adjuvant therapy with TNF-α on animal model of triple-negative breast cancer. Ther Deliv. 2018;9(5):333–42.

    Article  CAS  PubMed  Google Scholar 

  84. Shimizu K, et al. Enhancement of tumor lysate- and peptide-pulsed dendritic cell-based vaccines by the addition of foreign helper protein. Cancer Res. 2001;61(6):2618–24.

    CAS  PubMed  Google Scholar 

  85. Delneste Y, et al. Vasoactive intestinal peptide synergizes with TNF-α in inducing human dendritic cell maturation. J Immunol. 1999;163(6):3071–5.

    Article  CAS  PubMed  Google Scholar 

  86. Balkwill F. TNF-α in promotion and progression of cancer. Cancer Metastasis Rev. 2006;25(3):409–16.

    Article  CAS  PubMed  Google Scholar 

  87. Spitzer MH, et al. Systemic immunity is required for effective cancer immunotherapy. Cell. 2017;168(3):487-502.e15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Chen B, et al. Predicting HLA class II antigen presentation through integrated deep learning. Nat Biotechnol. 2019;37(11):1332–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Eiseman E, Bolen JB. Engagement of the high-affinity IgE receptor activates src protein-related tyrosine kinases. Nature. 1992;355(6355):78–80.

    Article  ADS  CAS  PubMed  Google Scholar 

  90. DiLillo DJ, Ravetch JV. Differential Fc-receptor engagement drives an anti-tumor vaccinal effect. Cell. 2015;161(5):1035–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Steinman RM, Dhodapkar M. Active immunization against cancer with dendritic cells: the near future. Int J Cancer. 2001;94(4):459–73.

    Article  CAS  PubMed  Google Scholar 

  92. Jonuleit H, et al. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol. 1997;27(12):3135–42.

    Article  CAS  PubMed  Google Scholar 

  93. Santana-Magal N, et al. Isolation protocol of mouse monocyte-derived dendritic cells and their subsequent in vitro activation with tumor immune complexes. J Vis Exp. 2018;135: e57188.

    Google Scholar 

  94. Koido S. Dendritic-tumor fusion cell-based cancer vaccines. Int J Mol Sci. 2016;17(6):828.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Currivan E, Finlay D, Moreira D. Dendritic cells metabolism: a strategic path to improve antitumoral DC vaccination. Clin Exp Immunol. 2022;208(2):193–201.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Santana-Magal N, et al. Melanoma-secreted lysosomes trigger monocyte-derived dendritic cell apoptosis and limit cancer immunotherapy. Cancer Res. 2020;80(10):1942–56.

    Article  CAS  PubMed  Google Scholar 

  97. Gutwillig A, et al. Transient cell-in-cell formation underlies tumor relapse and resistance to immunotherapy. Elife. 2022;11: e80315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Altorki NK, et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 2021;22(6):824–35.

    Article  CAS  PubMed  Google Scholar 

  99. Ho AY, et al. A phase 2 clinical trial assessing the efficacy and safety of pembrolizumab and radiotherapy in patients with metastatic triple-negative breast cancer. Cancer. 2020;126(4):850–60.

    Article  CAS  PubMed  Google Scholar 

  100. Hua Y, et al. Toripalimab plus intensity-modulated radiotherapy for recurrent nasopharyngeal carcinoma: an open-label single-arm, phase II trial. J Immunother Cancer. 2021;9(11): e003290.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Rasoulouniriana D, et al. A distinct subset of FcγRI-expressing Th1 cells exert antibody-mediated cytotoxic activity. J Clin Invest. 2019;129(10):4151–64.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Martin PJ, et al. Replication of associations between genetic polymorphisms and chronic graft-versus-host disease. Blood. 2016;128(20):2450–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Qasim W, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med. 2017;9(374): eaaj2013.

    Article  PubMed  Google Scholar 

  104. Stadtmauer EA, et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020;367(6481): eaba7365.

    Article  CAS  PubMed  Google Scholar 

  105. Wilhelm M, et al. Successful adoptive transfer and in vivo expansion of haploidentical γδ T cells. J Transl Med. 2014;12:45.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Leung W. Infusions of allogeneic natural killer cells as cancer therapy. Clin Cancer Res. 2014;20(13):3390–400.

    Article  CAS  PubMed  Google Scholar 

  107. Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–22.

    Article  CAS  PubMed  Google Scholar 

  108. Alnaggar M, et al. Allogenic Vγ9Vδ2 T cell as new potential immunotherapy drug for solid tumor: a case study for cholangiocarcinoma. J Immunother Cancer. 2019;7(1):36.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Seetharam A, Tiriveedhi V, Mohanakumar T. Alloimmunity and autoimmunity in chronic rejection. Curr Opin Organ Transplant. 2010;15(4):531–6.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (81872412, 81602303), the Basic and Applied Basic Research Funding of Guangdong Province, China (2023A1515010798), and the Open Projects of the Key Laboratory of Human Genetic Diseases Research of Inner Mongolia, China (YC202201).

Author information

Authors and Affiliations

Authors

Contributions

Authors contributed to this paper with the design (YL, YYH, and HWX), literature search (YL), drafting (YL), revision (YL, YYH, HWC, YYW, ZWM, YX, HYX, JQL and HWX), editing (YL, YYH, HWC, YYW, ZWM, YX, HYX, JQL and HWX) and final approval (YYH, HYX, JQL and HWX).

Corresponding authors

Correspondence to Hong-Yi Xin, Jun-Qing Liang or Hong-Wu Xin.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Huang, Y., Cui, HW. et al. Perspective view of allogeneic IgG tumor immunotherapy. Cancer Cell Int 24, 100 (2024). https://doi.org/10.1186/s12935-024-03290-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03290-9

Keywords