- 1Precision and Personalized Cancer Treatment Center, Division of Cancer Diagnosis & Therapy, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China
- 2Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical College, Xinxiang, China
- 3Division of Cellular & Biomedical Science, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China
Globally, in 2018, 4.8 million new patients have a diagnosis of gastrointestinal (GI) cancers, while 3.4 million people died of such disorders. GI malignancies are tightly relevant to 26% of the world-wide cancer incidence and occupies 35% of all cancer-associated deaths. In this article, we principally investigated molecular and cellular mechanisms of tumorigenesis in five major GI cancers occurring at esophagus, stomach, liver, pancreas, and colorectal region that illustrate high morbidity in Eastern and Western countries. Moreover, through this investigation, we not only emphasize importance of the tumor microenvironment in development and treatment of malignant tumors but also identify significance of M2PK, miRNAs, ctDNAs, circRNAs, and CTCs in early detection of GI cancers, as well as systematically evaluate contribution of personalized precision medicine including cellular immunotherapy, new antigen and vaccine therapy, and oncolytic virotherapy in treatment of GI cancers.
Introduction
Gastrointestinal (GI) malignant tumors generally cover esophagus, stomach, liver, pancreas, as well as colorectal region. Most primary cancers from those parts worldly covered 26% of cancer incidence and 35% of all deaths related to cancer, while 4.8 million new cases were yearly diagnosed and 3.4 million people lost their lives due to those malignancies, statistically in 2018 (1). Currently, clinical scientists are segmentally concentrated on contribution of the tumor microenvironment (TME) to tumorigenesis and resistance to anticancer therapy (2–4), feasibility of precise methodologies in detection of GI cancers at early stage based on tumor growth-associated serological and histochemical changes (5–12), as well as personalized application of biomedical therapies in reduction of tumor size and control of metastases (13–20). More importantly, through combating with cancers over several centuries, many clinicians eventually and principally come back to the nature of malignant tumor, in which an outside exhibition of malignant tumors is uncontrolledly growing but the inside mechanism shows primary inhibition of immune surveillance and cytotoxic activity locally and systematically.
Tumor Microenvironment and Tumorigenesis in Gastrointestinal Cancers
Tumor Microenvironment (TME)
Like Achilles, if we cannot lift him from the ground, he will be alive last long. The Achilles malignant tumors that depend on is the TME. The TME is more like a matrix, in which cancer cells communicate with non-transformed cells through a variety of cytokines, chemokines, growth factors, as well as inflammatory mediators, purposely to maintain a dynamic equilibrium in causing metastases and resisting to physical, chemical, and immune pressure (2, 21, 22). The following components principally play critical roles in tumorigenesis and metastasis, including dendrite cells (DCs), T lymphocyte, natural killer cells (NKs), tumor-associated macrophage cells (TAMs), cancer-associated fibroblast cells (CAFs), and myeloid-derived suppressor cells (MDSCs) as well as transform growth factor β (TGFβ).
Dendrite Cells (DCs)
DCs, as antigen-processing vehicles, are tightly communicating with tumor-infiltrating T cells in purpose of elimination of carcinogenic cells and prevention of local tumorigenesis and systematical metastases. Primarily, DCs are functional to activate CD4+ and CD8+ T cell via antigen presentation, in which immature DCs intake the antigen to promote DCs maturation (23–25). However, based on hypoxic and inflammatory conditions in the TME, T cells activated by tumor-associated antigens are much less produced due to reduction of DC maturation, accumulation of immature DCs, and specific DC-mediated impairment of T-cell response (26, 27). Thus utilizing immune checkpoint inhibitors (ICIs) not only decrease programmed cell death protein 1 (PD-1) activity on DCs but also increase DC activity promoted via amonophosphoryl lipid A produced by E. coli, both of which strongly improve efficiency and efficacy of cellular immunotherapy to GI cancers (28–34).
T Lymphocytes
T lymphocytes inside or around the TME are the majorly powerful enforce to fight with cancer cells, in which tumor-infiltrating lymphocytes (TILs) have been purposely used to treat GI cancers and TILs expressed in the TME are closely relevant to therapeutic efficacy (35–40). Generally, cytotoxic CD8+-T cells are antigen-processed and have ability to eliminate tumor cells, whereas CD4+ T helper 1 cells (Th1) activate cytotoxic T cells to secrete interleukin-2 (IL2), interferon gamma (IFNγ), and tumor necrosis factor (41). Thereby, the higher expression of cytotoxic CD8+-TIL on biopsy, the better prognosis whether with cancer treatment or not (35–39). However, regulatory CD4+ cells, such as Th2 and Th17 group, focus on promoting tumor growth (41). Furthermore, CD4+ T regulatory cells (Tregs) with FOXP3 and CD25 expression release IL10, TGF-β, and cytotoxic T-lymphocyte antigen 4 (CTLA4), which tremendously diminish immune surveillance (42, 43). Expression of Treg cells in biopsy represents a negative prognosis (43). Interestingly, in Hodgkin’s lymphoma, Treg cells inhibit malignant cell growing, which show a good prognosis (44–46). More interestingly, γδT cells stained in the TME directly destroy transformed cells and upregulate activity of DCs, cytotoxic T cells, and NK cells in most malignant tumors (47–50). Invertedly, in situ production of IL17 via γδT cells recruits MDSCs and increases program death protein-ligand 1 (PD-L1) expression that is beneficial for tumorigenesis and metastases (51, 52). Due to contradictory results, whether presentation of γδ TILs in the TME is an eligible biomarker to evaluate prognosis has always been investigated.
Natural Killer Cell (NKs)
NK cells, another category of TILs, are characterized with CD16dimCD56bright subset mostly in circulation, whereas with CD16brightCD56dim subset predominantly in mucosa, in which CD56bright NK cells have cytotoxic action directly on cancer cells independent of antigen presentation (53). Different from functionally killing tumor cells and cancer stem cells (CSCs) in the bloodstream, NK cells show reduced cytotoxicity on transformed cells in the TME with producing less IFNγ (54). Unsurprisingly, through combating with immune defense system, malignant cells also develop several tools to dodge NK cell-mediated elimination including activation of inhibitory receptor of NK cells and reduction of NK cell activity induced by platelets and TGFβ (55). However, due to NK-mediated induction of DC cells and increased activation of more NK cells via ICI treatment, the existence of NK cells in biopsy highly predicts a good prognosis in GI cancer therapy (54, 56–58).
Tumor-Associated Macrophage (TAMs)
Macrophage cells are essentially in regulation of individual immunity through activating adaptive immunity, healing the wound, and kicking off infectious effectors, in which M1 type macrophages with IL12brightIL10dim are majorly activated by IFNγ and lipopolysaccharide, whereas M2 type macrophages with IL12dimIL10bright are typically activated by glucocorticoid and IL4/13 (59). M1 macrophages produce higher level of IL12 and drive the recruitment of Th1, which promotes antitumor activity, while M2 macrophage-mediated high secretion of IL10 largely sends votes to tumorigenesis. Within the TME, cytokines and inflammatory agents predominantly polarize M1 macrophage to M2 subtype, which produces TAMs (60). There are several receptors commonly expressing TAMs, such as the mannose receptor and scavenger receptor class A, in which blocking scavenger receptor class A via monoclonal antibody amplifies cytotoxicity of NK cells to transformed cells in the TME (61). In addition to immunosuppression, TAMs also notably increase tumor angiogenesis, especially at hypoxic area or hypoxia-induced tumor necrosis region where oligonucleotide microarray of aggregated TAMs shows extensive activation of the transcriptional zones encoding angiogenic factors such as vascular endothelial growth factor and endothelin (62). Furthermore, TAMs are strictly relevant with tumor invasion and metastases, and largely prevention of chemotherapy-induced apoptosis in GI cancers (63–65). These discoveries firmly identify high expression of TAMs in biopsy as a poor signal of prognosis in cancer therapy.
Myeloid-Derived Suppressor Cells (MDSCs)
MDSCs have two subpopulations including monocytic group and granulocytic group that are characterized through expression of different kinds of membrane markers and immunosuppressive components (66). MDSCs exhibit two directions in promotion of tumorigenesis and metastases including accelerating tumor angiogenesis (67) and inactivating innate and adaptive immune function through producing high concentration of reactive oxygen and nitrogen species (68), slowing down amino acid-mediated T-cell activation and proliferation by less production of arginine (69) and L-cysteine (70) within the TME and reducing intratumoral migration of cytotoxic T cells by peroxynitrite-modified chemoattractant CCL2 (71), as well as enlarging Treg cell-dependent immunosuppression via IL10, TGFβ, IFNγ, and CD40-CD40L interactions (72, 73).
Impressively, MDSCs have a triple communication with DCs, NK cells, and TAMs. In the TME, MDSC-TAM complex significantly regulates production of IL10 and IL12, in which IL10-knockdown animal model verifies that MDSC-dependent IL10 level is the key factor of reducing IL12 production through influencing IL12 transcription (74). But cytokine- and inflammatory mediator-induced production of MDSC-TAM complex directly leads to high level of IL10 and low level of IL12 in the TME, which promotes recruitment of Treg cells and predominantly inactivates antitumor function of cytotoxic T cells and NK cells (75). Moreover, higher IL10 production also increases IL4 expression, which basically supports more transformation of TAMs (76). Recent investigation found that granulocytic MDSC-induced inflammation decreased NKG2D expression, an activating receptor of NK cells, which reduced cytotoxicity in the TME (77), whereas monocytic MDSCs amplified expression of activating ligand Rae1 on NK cells to increase killing activity through NKG2D, which subsequently lysed MDSCs (78). Furthermore, combining with ICIs and N803, an IL15 superagonist, NK cell therapy increased cytolysis of MDSCs in squamous cell carcinoma (79). Also, another case in treatment of head and neck squamous cell carcinoma reported that orally bioagent SX-682, a small-molecule inhibitor of CXCR1 and CXCR2, tremendously abolished MDSC aggregation and amplified activity of infiltrating NK cells in the TME (80). These investigations strongly indicate that MDSCs suppress NK cell-mediated elimination of cancer cells in the TME. Through studying effects of MDSCs on DCs in melanoma patients, the evidence showed that MDSCs decreased antigen presentation of DCs, inhibited incubation of immature DCs, and blocked recruitment of DCs to cytotoxic T cells, all of which were activated via IL23-mediated Th17 cell pathway (78, 81, 82). More evidences showed that IL23-mediated Th17 cell proliferation damaged adaptive and innate immunity and significantly promoted tumor metastases (78, 83). Therefore, MDSCs primarily enhance TAM-induced immunosuppression and selectively downregulate DC and NK cell activity in promotion of tumor growth and metastases.
Cancer-Associated Fibroblast Cells (CAFs)
Healing tissue damage with scar essentially needs participation of fibroblast cells, in which paracrine signals transform residential fibroblasts to myofibroblasts that produce TGFβ and α-smooth muscle actin (84, 85). However, in the TME, myofibroblast cells independent from mutations in cancer cells or the epithelial-mesenchymal transition (EMT) are primarily recognized as CAFs that are abundantly expressed in most malignant tumors (86). In purpose of constantly activating proliferation and metastases, CAFs produce various kinds of growth factors such as hepatocyte growth factor, fibroblast growth factor, and insulin-like growth factor 1 (87). CAF secretion partially activates expression of vascular endothelial growth factor that highly causes angiogenesis (88). Cytokines and chemokines produced by CAFs are functional on cytotoxic T cells, Treg cells, and macrophages, which cause both immune enhancement and immunosuppression (89). Differently, CAF-secreted IL6, CXCL9, and TGFβ reduce T cell response to cancer cells, especially cytotoxic T cells (90), whereas CAF-mediated production of CXCL12 (91) and inactivation of focal adhesion kinase in cancer cells (92) amplify antitumor effects through diminishing stromal fibroblast activation. Moreover, CAFs in the TME also contribute to building up the extracellular matrix (ECM) that consists of fibrovascular cores and remodeling enzymes for increasing tumor density to resist selective pressure (90, 93), while TGFβ-activated CAFs crucially stop activity of anticancer medicine (94, 95). Collectively, CAFs promote tumorigenesis and metastasis through modifying the ECM, utilizing growth factors for angiogenesis, and preventing drug access to reduce therapeutic effects.
Transform Growth Factor β (TGFβ)
TGFβ almost regulates all types of human cells and acts as a double agent in the TME (96, 97). Generally, TGFβ secretion is locally responsible to maintain homeostasis, and higher level of TGFβ created by blood platelet and stromal components is responsible for healing the injury and regeneration; however, in the TME, not only stromal cells produce TGFβ through paracrine but also malignant cells autocrinally generate more TGFβ (98). Being a cytokine, TGFβ activates signal transduction through Smad-dependent or Smad-independent pathways, in which Smad-dependent signaling demonstrates that TGFβ-mediated phosphorylation of Smad family protein, Smad2 and Smad3, via TβR1 receptor interplays with the cofactor to form the complex that is subsequently translocated into the nucleus and binds with Smad binding element to activate gene transcription (99, 100); however, to Smad-independent signaling, TGFβ activity is amplified mostly through mitogen-activated protein kinase (MAPK) pathway that is activated via p38, c-Jun amino terminal kinase, extracellular signal-regulated kinase, and nuclear factor-κB as well as phosphatidylinositol 3 kinase-Akt (PI3K-Akt) (101, 102). The dual roles of TGFβ showing at early and advanced stage of cancers are not due to the change of TGFβ structure, but are mediated through mutant TGFβ receptors as well as interaction of cancer cell-secreted TGFβ with the whole stromal cells such as dendritic cells, T cells, NK cells, TAMs, CAFs, and MDSCs. To suppress tumor growth, TGFβ arrests cell cycle through inhibiting cyclin dependent kinase and c-Myc pathways (103), induces cell apoptosis through activating JNK and Fas pathway, and antagonizing Bcl-2, Bcl-X, and survivin (104), and eventually prevents cell immortalization through regulating reverse transcriptase activity of human telomerase (105). To support tumorigenesis and metastases, paracrinal and autocrinal TGFβ enhances immunosuppression through diminishing immune surveillance maintained by DCs, cytotoxic T cells, and NK cells as well as cytokines such as IL2 and INFγ (22), amplifies angiogenesis through activation of ALK1 and ALK5 pathway and more production of vascular endothelial growth factor and connective tissue growth factors in epithelial cells and fibroblasts (97), and induces the EMT for promoting metastases through Smad-dependent and Smad-independent pathway (98), as well as accelerates malignant cells invasiveness through remodeling the ECM and decreasing TβR2 signaling and miRNA-mediated protein regulation (99).
Furthermore, hyperactivation of TGFβ signaling in advanced-stage malignant tumors also swiftly decreases efficacy of ICI therapy in the TME (106), whereas malignant tissue insensitive to ICI treatment exhibits a high expression pattern of TGFβ1 (107). Neutralized monoclonal antibodies individually targeting on TGFβ or dual recognition of TGFβ and PD-1/PDL-1 have achieved significant antitumor effects in treatment of liver cancer, pancreatic cancer, lung cancer, and urethral cancer (108, 109). Therefore, once malignant cells finish the transition from locally seeding to angiogenesis, TGFβ ultimately abandons the antitumor arm and majorly serves to immunosuppression and metastases.
Tumorigenesis in Gastrointestinal Cancers
Esophageal Cancer
Esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC) are two major malignant exhibitions at esophagus. Epidemiological studies demonstrate that multiple factors cause EAC and ESCC, such as smoking, obesity, gastroesophageal reflex disorder, and Barrett’s esophagus, as well as epithelial injury via frequently intaking hot solution, all of which indicate that chronic inflammation is mainly responsible for malignant transition (110). In chronic inflammation-mediated esophageal cancers, not only IL6-STAT3 (signal transducer and activator of transcription 3) pathway increases epithelial proliferation and apoptotic resistance in Barrett’s esophagus and EAC but also neoplastic cells utilize STAT3 signaling to promote tumorigenesis, angiogenesis, and metastases (111, 112). To ESCC, increased production of IL6 causes a poor prognosis following neoadjuvant radiochemotherapy, while higher expression of STAT3 at surgical resection correlates to increased mortality; however, siRNA-activated inhibition of IL6/STAT3 signaling augments chemotherapy sensitivity, accelerates apoptosis, and reduces angiogenesis, as well as decreases the EMT formation for metastases (113, 114).
More importantly, in EAC and ESCC, high expression of CD38-positive MDSCs is abundantly found in the peripheral blood at advanced stage, while Daratumumab binding with CD38 expression reduces tumor growth (115). Previous studies reported that MDSCs increased recruitment of Treg cells in maintenance of immunosuppression in the TME (116). It is highly possible that MDSCs promote malignant invasion through increasing Treg cell activity, in which the higher level of Treg cells, the stronger prediction of tumor invasion, metastasis, disease severity, and reduced overall survival (114, 117). Furthermore, due to the TME in esophageal cancers, especially in ESCC, enormously supporting transition of M1 macrophages to TAMs, infiltrating TAM-based immunosuppression (113), and TAM-mediated upregulation of Treg cell activity tremendously promote the development of EAC and ESCC through STAT3 signaling (118). Additionally, TGFβ directly increases CAF production in the TME, because of which, CAF-based secretion of vascular growth factors accelerates metastases in ESCC, whereas inhibition of CAF activity in EAC notably improves overall survival associated with an excellent prognosis (119). Collectively, interplay of various chronic inflammation conditions activates tumorigenesis in esophageal cancer.
Gastric Cancer
Similar to esophageal cancers, most environmental issues are directly and indirectly functional on generating gastric malignancy including high-salt food, pickled food, smoking, nitrates, and food preservers (120). But malignant tumors occurring at cardia and gastroesophageal junction are mostly related to gastroesophageal reflux, intestinal mucosa transition, and chronic mucosal infection caused by H. pylori (Hp) (121). External factors and internal issues confirm the contribution of chronic inflammation in promotion of gastric cancer (122, 123). Two pathological categories in gastric cancer are intestinal type and diffuse type, in which intestinal type is closely related to Hp infection in older males and less morbidity in last several decades worldly, while diffuse type is characterized with loss of E-cadherin expression, frequently occurring in young people unisexually, and less correlating with precancerous lesion (124).
As extensively reported, microorganisms make a great contribution in stimulation of tumorigenesis through dysregulation of cycle progression, protein translation, and cell survival. Epstein Barr Virus (EBV) and Hp are frequently emphasized due to mostly triggering PI3K-Akt pathway and Wnt/β-catenin pathway, respectively (121, 125). Crucially, mutant PI3KAc, an oncogene encoding a catalytic subunit of PI3Kα, is remarkably expressed in up to 42% EBV-associated gastric adenocarcinoma, which predominantly causes DNA hypermethylation, especially promoting tumor suppressive gene CDKN2A hypermethylation (125). Moreover, amplification of mutant PI3KAc swiftly increases angiogenesis stimulated by VEGF and EGF through TGFβ-CAFs pathway (125). Also blocking oncosuppressive gene, PTEN, activates Akt expression and phosphorylation of Akt, which strongly dysregulates cell apoptosis, tumor invasiveness, and radiosensitivity (126). miRNA-21 targeting on PTEN expression rises proliferation and invasiveness of gastric cancer (126). Dysfunction of Wnt/β-catenin pathway occurs in about 70% gastric cancer patients, in which two virulence factors of Hp, CagA and VacA, are mainly responsible for such dysregulation (127). CagA not only interplays with E-cadherin to increase β-catenin accumulation in cytoplasm and nucleus but also activates CDX1 and P21 genes also firmly responsible for transition of Hp-infected gastric epithelium to mesenchymal stemness such as CSCs (128). Interestingly, chronic inflammation induced via Hp infection reprograms SALL4 expression, which directly promotes intestinal metaplasia to neoplastic transformation (129), whereas increased interaction of SALL4 with Wnt/β-catenin signaling also accelerates lymphatic node metastases in gastric cancer patients (130). Furthermore, unsurprisingly, TGFβ is a valid sponsor in upregulation of Hp-induced EMT (131).
Activation of EGFR is responsible for up to 30% gastric cancer cases, in which increased EGFR expression or mutant EGFR is responsible for 5–10% patients and correlates with microsatellite instability molecular subtypes (132). Various coregulators binding with EGFR and TNFα recruit signaling cascades that activate MAPK and PI3K-Akt pathways (133). Nevertheless, HER-2 overexpression exists in 10–30% gastric cancer patients, in which mutant HER-2 occurs in 5% gastric cancer cases, and is more prevalent in 7% microsatellite instability cases and 12% EBV-positive ones (132). Thereby, overexpression of mutant EGFR and HER-2 is highly associated with metastases and poor prognosis in gastric cancer patients (132). Furthermore, specific categories of miRNAs functionally contribute to the development of gastric cancer through increased oncogene expression, reduced expression of oncosuppressive genes, and dysfunction of tumor suppressive proteins; therefore, depending on miRNAs, they are highly likely to become a practically clinical tool in detection of GI cancers and evaluation of prognosis (134).
Liver Cancer
Hepatocellular carcinoma (HCC) mostly is diagnosed at the late stage following viral infection, alcohol overconsumption, steatohepatitis, primary biliary cirrhosis, autoimmune hepatitis, and aflatoxin contamination, in which chronic viral infections, such as HBV and HCV, are responsible for 80% cases (135). More researchers believe that oncogenic background changes, dysregulation of up- and downstream molecular pathways, and interaction between stromal cells and cytokines in the TME are tightly connected with cancer cell transition, proliferation, angiogenesis, invasion, and metastasis in HCC.
Single-cell transcriptomic profiling of HCC patients identifies that TME reprogram mainly controls HCC biodiversity, and VEGF-A-mediated regulation of CAFs and TAMs activities is only exhibited in malignant cells, especially in high diversity tumors, but not found in non-malignant hepatocytes (136). This investigation also verifies that individual activation of VEGF-A pathway in HCC is completely responsible to immunosuppression, aggressiveness, and negative prognosis (136). Also, VEGF-A is a critical trigger of angiogenesis in HCC serving for invasion and metastases (137). In HBV- and HCV-mediated hepatitis, chronic infection leads to cirrhotic situation that causes a hypoxic environment, in which hepatocytes upregulate hypoxia-induced factor 1α expression, and consequently that causes overexpression of VEGF at transcriptional level, and finally promotes angiogenesis in HCC (138). Treatment using lenvatinib and sorafenib targeting on VEGFs notably significantly improves prognosis in HCC patients (139). As mentioned previously, TGFβ dysregulated VEGF expression in different kinds of cancers (99); however, TGFβ is a double agent in health and diseased liver. In health liver, Kupffer cells and stellate cells produce most TGFβ except for hepatocytes, while in an injured health liver, temporary activation of TGFβ is functional on nuclear Yes-associated protein and Smad2 phosphorylation (140) that induces the EMT formation dependent on activated hepatic stellate cells and hepatocytes-transformed myofibroblast cells. Moreover, stromal cells in the TME utilize the fibrosis process induced by EMT for metastases, which has been actually verified through miRNA-dependent inactivation of TGFβ pathway in reducing HCC tumorigenesis and improving prognosis (141, 142).
More importantly, overexpression of EGFR and insulin-like growth factor receptors enhances activation of PI3K/Akt/mTOR signaling in HCC, in which atypical PTEN activity upregulates activation of the PI3K/AKT/mTOR pathway (143). Previous researches demonstrated that mutant PTENTs were identified in 5% HCC patients, while reduced or deleted PTEN expression was associated with nearly 50% HCC cases (144, 145). Abnormal expression of PTEN in chronic HBV- and HCV-infected patients constantly stimulates overactivation of PI3K/AKT/mTOR pathway, which constitutively promotes tumor grades and advanced disease stage, and diminishes overall survival in HCC patients (144). Clinically, everolimus and mTOR inhibitor plus a PI3K inhibitor (BKM120), Akt inhibitor (MK-2206), and mTOR/PI3K inhibitor (BEZ235) significantly increase immune cytotoxic activity, especially in treatment of advanced HCC via everolimus, in which 40–73% patients quickly catch the disease stable state in a short-time study (146, 147).
Alternatively, in almost 50–70% HCC patients, immunostainings identify overexpression of β-actin in cytoplasm and nucleus especially for tumor cell proliferation and suppression of physiological differentiation; however, under nonmalignant situation, high accumulation of β-actin shows less contribution to HCC formation (148). Furthermore, the early-stage HCC demonstrates that β-actin mutation occurs in 17% HCC patients and these mutant regions cover some specific sites such as phosphorylated site of glycogen synthase kinase-3β (149). Investigation of clustering β-actin resource in HCC shows that mutant β-actins are responsible for 12–26% of patients, whereas mutations in Wnt/β-actin pathway contribute 8–13% cases, both of which predominantly occur in HCV-infected population (149). Clinically, genetic profiling of 603 HCC patients illustrates that either Wnt/β-actin pathway or Myc/Akt pathway is abnormally activated, which is parallel with TGFβ overexpression (150). Interestingly, through activation of Wnt/β-actin pathway, miRNAs regulate TGFβ-mediated ECM formation (151), and more evidences expose that due to the ECM transition, liver has generally become a common site not only for HCC metastasis but also for other solid tumors (152, 153).
Pancreatic Cancer
As a deadly disease, pancreatic cancer, like mountain Everest, has always been attractive to many physicians and surgeons in that 5-year survival rate is only 8% (154), in which 90% pancreatic cancer is pathologically defined as pancreatic duct adenocarcinoma (PDAC) and 10-year survival rate is less 4% (155, 156). Principally, in pancreatic cancer, the TME promotes tumor growth and angiogenesis, CSCs focus on anticancer therapy and increased tumor bulk, and interaction between CSCs and the EMT contributes to metastases.
One of pathological characteristics in PDAC is desmoplasia, a state of extensive fibrosis around the primary tumor region, in which the ECM proteins are overexpressed and myofibroblast cells are tremendously identified (157). Clinical evidences also demonstrate that desmoplasia displaying in lymphonodes of the PDAC patients predicts a poor prognosis (157). Desmoplasia occurring in PDAC tightly depends on the TME in that TME-associated secretion of connective tissue growth factors, fibroblast growth factor 2, and TGFβ stimulates desmoplasia formation, especially the absence of anticancer arm of TGFβ caused by reduced Smad4 function depending on RAS-mediated ERK pathway (158). Due to high fibrosis in desmoplasia, the TME exhibits hypovascular and hypoxic conditions, through which pancreatic cancer cells reprogram metabolic pathways, inactivate apoptosis, and amplify proliferation and anticancer resistance, as well as promote invasion and metastasis (159). In purpose of maintenance and enhancement of hypoxia, pancreatic cancer cells produce more angiostatin, endostatin, and pigment epithelium-derived factors, whereas the ECM creates more endostatin, both of which are eventually countering vessel growing (160, 161). Desmoplasia-induced hypoxia, especially via hypoxia-induced factor 1α and TGFβ, constantly stimulates production of activated pancreatic stellate cells, in which stellate cells with quiescent vitamin-A contents transform into myofibroblast-like type (162). Through this procedure, normal cellular framework in pancreatic microenvironment steps into the carcinogenic ECM, which is responsible for upregulating secretion of cytokines and chemokines such as matrix metalloproteinases, platelet-derived growth factor, epidermal growth factor, insulin-like growth factor 1, fibroblast growth factor, and stromal-derived factor 1, as well as small leucine-rich proteoglycans and collagen type I, all of which make a great contribution to cancer development and metastasis (162, 163). Moreover, activated pancreatic stellate cells notably participate into immunosuppression in the TME, in which activation of stellate cell-dependent IL6 secretion causes MDSC aggregation deliberately to reduce T-cell proliferation, and increases Treg cell infiltration via STAT3 pathway (164). In addition, activated stellate cells also cause more T-cell apoptosis and Th2 cytokine secretion in the TME through upregulating Galectin-1 expression (165). Recently, investigation identified that inhibiting heat-sensitive protein 90 restricted transformation of cancerous pancreatic stellate cells and diminished IL6 production in the TME, which significantly amplified effects of ICIs and increased cytotoxic T cells and Th1 cell infiltration through inactivation of JAK/STAT and MAPK signaling in PDAC (166).
Hypoxia in PDAC not only stimulates production of activated stellate cells but also promotes stemness, in which hypoxic environment indirectly and directly supports CSC creation (167, 168). Growing with pancreatic cancer cells in vitro simultaneously, activated stellate cells increase spheroid formation of malignant cells and upregulate expression of stemness markers such as ABCG2, Nestin, and LIN28, which strongly indicates the importance of activated stellate cells in generating CSC niches (169). Specially, TGFβ1 predominantly secreted by activated stellate cells in PDAC stimulates CSC production through negatively regulating L1 cell adhesion molecule expression by TGFβ-Smad2/3 pathway, which causes PDAC to be more progressive (170). More importantly, self-renewal CSCs have become a major barrier of conventional chemoradiotherapy, in which gemcitabine is unable to eradicate CSCs but rather increase the number of CSCs (171, 172). In purpose of maintaining the renewal, increasing the tumor bulk, and promoting metastases, CSCs dysregulate several signal pathways including Wnt/β-catenin, hedgehog, notch, NF-κB, PI3K/Akt, and PTEN (173). Unsurprisingly, CSCs also secret Actin and Nodal that interact with Alk4/7 receptors to form pancreatic cancer cell sphere; however, through reduction of Akl4/7 expression and inhibition of interplay with CSCs, pancreatic cancer cells are more sensitive to gemcitabine treatment and lead to a long overall survival (174).
For entirely understanding the procedure of metastases in PDAC as well as contribution of CSCs in metastases, more investigations evidently show that the EMT insidiously plays a pivotal role during invasion through inducing CSC production continuously (175, 176). The most critical event in forming the EMT is to transform epithelial cells from a normally mature condition into mesenchymal phenotype, which causes morphogenetic changes occurring in embryonic development, organ fibrosis, and tumor metastasis, as well as less expression of epithelial markers such as E-cadherin, desmoplakin, and plakoglobin and overexpression of mesenchymal markers including vimentin, fibronectin, and α-smooth muscle actin (177, 178). Interestingly, hypoxia-inducible factor 1α is functional on expression of SIP1, Snail, Twist1, and Zeb1 indirectly or directly, while hypoxia-inducible factor 2α directly regulates Twist1 expression, both of which enlighten the essential role of hypoxia in induction of the EMT formation (179–181). Impressively, Zeb1 suppresses E-cadherin expression to promote metastases through downregulating expression of stemness inhibitors such as miR-203 and miR-200, in which TGFβ directly causes upregulation of Zeb1 signaling in the EMT (176, 182). Furthermore, circulating tumor cells (CTCs) depending on the EMT-mediated extravascular invasion and migration occupy 100-fold increases of CD24+ and CD44+ expression that are highly exhibited in pancreatic CSCs associated with therapeutical resistance and negative prognosis (183, 184). That TGFβ predominantly secreted by activated stellate cells strongly enhances CSC production in desmoplasia indicates that interaction between CSCs and the EMT crucially supports metastatic infrastructure and pathological progression in PDAC.
Colorectal Cancer
Clinical practice using non-steroid anti-inflammable drugs such as COX-2 antagonists but not aspirin exhibits 40–50% reduction at risk of colorectal cancer (CRC), which strongly demonstrates that chronic inflammation initially promotes tumorigenesis in CRC (185, 186). Further evidences verify that COX-2-mediated prostaglandin E2 production is critically responsible for CRC invasion (187). However, recent researches substantially identified that prostaglandin E2 secreted by CAFs essentially correlates with immunosuppression, angiogenesis, and metastases in CRC, in which upregulation of COX-2 expression on CAFs promotes migration and invasiveness (188), reduction of COX-2-mediated miR-335 expression blocks CAF-induced carcinogenesis through restoring PTEN activity (189), and dysregulation of NK cell function by hepatocellular CAFs depends on prostaglandin E2 and indoleamine 2,3-dioxygenase activity (190). Therefore, transformation of normal fibroblasts to CAFs is a milestone in CRC formation, which purposely remodels the TME through reprogramming secretion of the ECM proteins (191). Moreover, CAFs in CRC strongly amplify immunosuppression in the TME through CAF-mediated enhancement of TAM and MDSC activities (192, 193), CAF-dependent promotion of Treg cell proliferation (194), and CAF-induced PDL-1 overexpression on cancer cells (195), as well as CAF-activated reduction of cytotoxic T cell activity (196). Furthermore, exosomes predominantly secreted by CAFs typically contain miR-21, miR-378e, and miR-143, most of which travel from CAFs to cancer cells in response to increased expression of the EMT markers and stemness to become more aggressive phenotype, which is parallel with results obtained from cancer cells transfected with miR-21, miR-378e, and miR-143 (197). Therefore, miR-21-contained vehicles majorly created by CAFs play an essential role in upregulation of cancer cell-based tissue damaging, especially causing more metastases to liver in CRC (198). Eventually, CAF-dependent exosome secretion is inherently in recruitment of CSCs to resist oxaliplatin or 5-fluorouracil treatment in CRC therapy; however, inhibition of exosome production significantly reduces such kind of behaviors (199). In addition, CAF-induced IL17a secretion also notably promotes self-renewal and invasiveness of CSCs and worthfully to know that chemotherapy of CRC unfortunately upregulates IL17a secretion (200).
Clinical Approaches in Diagnosis of Gastrointestinal Cancers at Early Stage
Currently, conventional screening cancer tools including plasma-based tumor markers, barium enema, GI endoscopy, and computed tomography have been practiced extensively in asymptomatic population for decades of years in purpose of identifying malignant tumors at early stage, in which GI endoscopy is strongly recommended due to high accuracy of histology; however, invasive nature, unhappier feeling, and noticeable cost have conditionally slabbed this optimal practice for a large-scale of screening tests (201, 202). Most of serum tumor markers generally show less specific in early detection of GI malignancies except for αfetoprotein and CA19-9 with highly predictable value of hepatocellular cancer and pancreatic cancer, respectively (203, 204). According to diversity of cytokines, chemokines, and glycoproteins in the TME, it is necessary to comprehensively apply novel biomarkers for early diagnosis of GI malignancies, which are not independent from traditional cancer-screening tests.
M2 Pyruvate Kinase (M2PK)
Glutaminolysis and glycolysis almost exist in all kinds of solid tumors in that hypoxia in the TME significantly upregulates glycolysis d in malignant tissues (205, 206). Being a rate-limiting enzyme, M2PK plays a crucial role in regulation of glycolysis, in which tetrameric structure of M2PK exhibits high affinity to phosphoenolpyruvate, whereas a dimeric form, named tumor M2PK with low affinity to phosphoenolpyruvate, is predominantly expressed in malignant tumors (207). A large number of evidences demonstrate that increased expression of tumor M2PK in screening samples especially correlates with GI cancers and also has a pan-sensitive to malignancies in brain, thyroid, lung, breast, ovary, cervix, kidney, bladder, and prostate (205, 208–215).
As a meta-analysis study showed, tumor M2PK as a plasma marker had 62.1% sensitivity and 89% specificity in detection of gastric cancer and esophageal cancer (216). According to different stages of gastric cancer, tumor M2PK has 71% sensitivity to metastatic cases, whereas CA72-4 is 57%, while to the cases without metastases, tumor M2PK has 63% sensitivity, whereas CA72-4 is 25% (11). The study focusing on accuracy of tumor M2PK, CEA, CA19-9, and CA72-4 in diagnosis of GI cancers confirms that tumor M2PK is significantly higher in screening esophageal cancer, gastric cancer, and CRC than other three tumor markers, while in pancreatic cancer, tumor M2PK has a similar predictable rate as CA19-9, but more sensitive than CEA and CA72-4 (6). Furthermore, a comparable meta-analysis study found that screening pancreatic cancer through CA19-9 and M2PK together has 60% sensitivity and 95% specificity (11). More conveniently, testing tumor M2PK in fecal samples is significantly sensitive and reliable than fecal occult blood test (FOBT) due to intermittent bleeding or the absence of bleeding in CRC, in which tumor M2PK shows 68.8 to 91.0% sensitivity and 71.9 to 100% specificity, whereas FOBT has 40% sensitivity only or less (217). Therefore, combining with conventional plasma tumor markers and periodic endoscopy examination, tumor M2PK is practically selected as an earlier alert signal in detection of GI malignancies, especially in screening CRC.
Circulating Tumor Cell and Circulating Tumor DNA
CTCs are tumor cells that have broken away from primary tumors or metastatic sites, and then enter into the blood circulation in purpose of distal metastases (218). Comparing with other peripheral blood cells, the number of CTCs is extremely low, generally 10-7 to 10-8, and lifetime mostly lasts for several hours (219–221). Currently, using CTCs to screen GI cancers in normal population is not actually realistic due to remarkably low fraction of CTC-positive patients, screening markers less specific for CTCs assessment, and tremendously lower level of CTCs detected at early-stage tumors (222–224). A cohort study of 138 CRC patients at stage I to III demonstrates that postoperative CTC-positive patients who were negative at preoperative CTC test independently predict a really poor prognosis, whereas preoperative CTC-positive patients have similar results as preoperative CTC-negative group in evaluation of prognosis (225). Therefore, CTC test may be highly valid to patients with extensive metastases, post-surgery relapse, or the high-risk population with family history of GI cancers as well as a good candidate in prediction of prognosis after chemoradiotherapy.
Circulating tumor DNA (ctDNA) is a kind of cell-free DNA originally traveling from primary cancer site, metastatic location, or CTCs to the bloodstream, in which the size of ctDNAs is generally shorter than normal cell-free DNAs, lifetime lasts for 16–150 min, and can be released into exosomes (226, 227). Different from CTC test, ctDNAs screening has employed tumor-specific genetic markers and epigenetic markers in discrimination of ctDNAs from normal cell-free DNAs (228), and sensitivity of ctDNA detection has exhibited microscale level through molecular barcoding techniques (229). Furthermore, identification of ctDNAs through low-coverage genome sequencing has not required genomic information of primary malignancy (230), and ctDNA detection using plasma samples is more accurate than in serum due to lower level of wild-type DNA released (231). Through combinative application of both ctDNAs and CTCs techniques as well as through detection of hypomethylation or hypermethylation of ctDNAs at promoter region of cancerous and oncosuppressive genes, ctDNAs have been extensively used in early diagnosis of GI cancers. However, due to few amounts of ctDNAs released into the circulation, ctDNA technique is encountering some challenges in detection of ultra-early stage of GI malignancies (232, 233).
In esophageal cancer, ctDNAs targeting CASZ1, CDH13, and ING2 genes show significant high level of hypermethylation in ESCC patients but mild level in healthy controls, whereas dysregulation of 5-hydroxymethylcytosine expression on ctDNAs also has remarkable sensitivity in screening ESCC; therefore, ctDNA detection based on these two different patterns is highly likely to identify the early-stage esophageal cancer (234, 235). Through screening methylation and hypermethylation of 14 genes in 1193 samples, ctDNA approach has 65% sensitivity and 95% specificity in diagnosis of gastric cancer, whereas in comparison with CEA, CA72-4, CA19-9, and CA50, the higher concentration of ctDNAs tested by alu-dependent branch DNA assessment, the more accuracy in early diagnosis of gastric cancer, which has 79% sensitivity and 91.8% specificity (236, 237). Therefore, combining hypermethylation of CASZ1, CDH13, and ING2 genes with increased ctDNA burden is a valuable approach for a large-scale screening of upper digestive cancers. Using somatic copy number aberration (SCNA) via low-depth whole genome sequencing technique demonstrates that increased ctDNA burden has area under curve (AUC) value of 0.874 in early diagnosis of HCC and 0.933 at advanced stage of HCC, whereas application of ctDNA panel focusing on eight targeted genes has 83.3% sensitivity and 90.5% specificity in screening HCC; thereby, screening people through a panel containing multiple ctDNAs with increased ctDNA burden comprehensively provides a specific method in diagnosis of HCC (238, 239). To detect pancreatic cancer, several investigations clarify that single ctDNA-mediated tests are not sensitive at the early-stage; however, through combining with conventional serum tumor markers, specific proteins expressed in the TME and methylation at protomer regions of cancer-related genes, ctDNA technique significantly increases sensitivity and specificity in diagnosis of pancreatic cancer (240, 241). Using ctDNAs to detect methylation expression at promoter regions of 17 genes exhibits 91.2% sensitivity and 90.8% specificity to discriminate PDAC patients from chronic pancreatitis group (242). Through examining methylation at both ADAMTS1 and BNC1 genes in screening PDAC, ctDNAs tests remarkably accomplish the accuracy with 94.8% sensitivity and 91.6% specificity (243). Moreover, detection of mutant KRAS through ctDNAs combined with other four protein markers notably has 64% sensitivity and 99.5% specificity in diagnosis of PDAC patients from healthy controls (244). Thereby, the methodology of integrating ctDNAs with KRAS mutation, methylation on targeted genes, and high expression level of tumor-specific proteins predominantly occupies a position in early diagnosis of pancreatic cancers. Similarly to detect early gastric cancer, increased ctDNA burden through Alu83 and Alu244 is also used to detect CRC, combination of which with methylation of specific gene achieved higher sensitivity in CRC diagnosis (245). Furthermore, using ctDNAs to examine B4GALT1 gene hypermethylation performs 50% sensitivity and 100% specificity in early diagnosis of CRC, whereas hypomethylation of LINE-1 gene tested by ctDNAs yields 65.8% sensitivity and 90.0% specificity in screening CRC (246). Therefore, testing M2PK-positive samples depending on increased ctDNA burden, hypermethylated B4GALT1 gene, and hypomethylated LINE-1 gene is a reliable methodology in early detection of CRC.
Circulating microRNA
microRNAs (miRNAs) generally composing of 19–24 nucleotides are functional on 3’-UTR of messenger RNA, which effectively regulates targeted protein expression (247). miRNAs are endurable in various physiological changes and resist to RNase activity, thereby circulating miRNAs extensively and stably present in 12 kinds of cell-free body fluids and excretions including serum, plasma, urine, and feces (248). Since downregulation of miR-15 and miR-16 expression was firstly recognized as a novel biomarker in diagnosis of B-cell chronic lymphocytic leukemia (249), clinical professionals have applied circulating miRNAs in detection of GI cancers considerably and identified several unique expression patterns in early diagnosis of esophageal cancer (250, 251), gastric cancer (252), liver cancer (253), pancreatic cancer (254), and CRC (255).
Comparing to healthy controls, the expression panel containing miR-16-5p, miR-197-5p, miR451a, and miR-92a-3p is specially associated with ESCC, whereas the group of miR-16-5p, miR-320c, miR-638, and miR-92a-3p is significantly higher in squamous dysplasia, a precancer pathology, especially miRNA-21 overexpression highly correlating with alcohol-induced ESCC (250, 251). In a cost-effective screening of gastric cancer, 12-miRNA panel shows 87% sensitivity and 68.4% specificity, which is remarkably higher than CEA and CA-19-9 (252). A meta-analysis study of circulating miRNA-mediated detection of HCC demonstrates that the higher expression group of circulating miR-21, miR-122, and miR-223 is more specific in diagnosis of HCC than in healthy, hepatitis, or cirrhosis group, in which confident rates of miR-21, miR-122, and miR-223 are 0.9293, 0.8128, and 0.8597, respectively, especially miR-21 expression in high priority (256). Screening of pancreatic cancer proves that the expression group containing miR-125a-3p, miR-5100, and miR-642b-3p achieves the most promising result in discrimination of cancer patients from healthy ones, which shows 98% sensitivity and 97% specificity (254). Moreover, in diagnosis of CRC, the expression pattern consisting of miR-15b, miR-17, miR-21, miR-26b, and miR-145 has the best predictable performance, especially miR-21 and miR-26b with maximal specificity (257). Recently, circulating exosomes were also considered as the early biomarkers in diagnosis of GI cancers; however, the reliability of this approach mainly depends on analysis of contents within the circulating exosomes, especially miRNAs and cell-free DNAs (258).
Finally, through reviewing 42 investigations significantly relevant to the diagnostic performance of circulating miRNAs in GI cancers, this meta-analysis study concludes that comparing to CEA and CA19-9, circulating miRNAs have become reliable biomarkers in early diagnosis, moderately with 75% sensitivity and 81% specificity, and multiple-miRNA screening assay significantly achieves more accurate result than a single-miRNA test, as well as plasma-dependent miRNAs assay is precisely used in diagnosis of gastric cancer, while serum-based miRNA test is more suitable for CRC detection (259).
Circular RNA
Circular RNAs (circRNA), non-coding RNA with closed circular form, have been discovered in mammalian cells for decades and structurally characterized through high-throughput sequencing over 10 years, in which circRNAs are functional on the axle of circRNA-miRNA-mRNA responsible for target gene expression, interplay with RNA-binding protein in regulation of target protein activities, and act as posttranscriptional regulators influencing parental gene transcription and splicing (260). Due to strongly resisting to exoribonuclease, circulating circRNAs are stably and extensively expressed in exosomes, serum, plasma, saliva, and urine (261). Since ciRS-7 firstly exhibited sponging action on miRNA-7 to promote carcinogenesis, different kinds of cancers have shown unique existing patterns of circRNAs that are practically applied in diagnosis of malignancy at early stage (261). Moreover, recent investigation demonstrated that artificial expression of circRNA targeting on miRNA21 eliminated miRNA-21-mediated promotion of gastric cancer (262). Therefore, examining expression pattern of circRNAs sponging on cancer-specific miRNAs is highly likely to become a more efficient methodology in early diagnosis of GI malignancies.
Using a pool of circRNAs to screen ESCC identifies that upregulation of circ-DLG1 and circ-TTC17 combining with downregulation of hsa_circ_0001946, hsa_circ_0062459 and circ-SMAD7 has 79% sensitivity and 85% specificity, in which positive samples in this pool test have 5-fold higher possibility in transition to ESCC than normal controls (263). Through analyzing 343 plasma circRNAs expressed in gastric cancer patients and healthy controls, downregulation of hsa_circ_0001017 and hsa_circ_0061276 has the best diagnostic performance with 95.5% sensitivity and 95.7% specificity (264). Another study shows that lower expression of plasma hsa_circ_0000181 has 99.0% sensitivity and 85.2% specificity in gastric cancer (265). Therefore, the expression group of hsa_circ_0001017 and hsa_circ_0061276 and hsa_circ_0000181 is practically becoming a novel biomarker for early diagnosis of gastric cancer. For HCC detection, upregulation of seven circRNAs expression plus downregulation of five circRNAs expression in serum shows the best combinative performance, in which the expression group covering hsa_circ_0004001, hsa_circ_0004123, and hsa_circ_0075792 has 90.5% sensitivity and 78.1% specificity (266). Furthermore, two independent studies identically discover that plasma circ-LDLRAD3 overexpression significantly correlates with pancreatic cancer, in which together with CA19-9, combinative test has 80.3% sensitivity and 93.6% specificity in early diagnosis of pancreatic cancer (267, 268). Thus, integrating conventional tumor markers with circRNA expression highly possibly obtains more precise diagnosis in screening of pancreatic cancer. Similar methodologies are also used in early diagnosis of CRC, in which combining with CEA and CA19-9, the group of downregulated expression of plasma circ-CCDC66, circ-ABCC1, and circ-STIL increases AUC value 0.780–0.855 (269). Another investigation focusing on early diagnosis of CRC shows that using upregulated expression group of hsa_circ_0001900, hsa_circ_0001178, and hsa_circ_0005927 precisely identifies CRC patients in the CEA-negative group with AUC value 0.859 (270). Therefore, combinative patterns of using circRNAs with CEA and M2PK will maximally increase the diagnostic accuracy in screening CRC.
Cellular Immunotherapy in Treatment of Gastrointestinal Cancers
Cellular immunotherapy is a combinative methodology of applying cellular therapy and immunotherapy in personalized cancer treatment (271–273), in which personalized analysis to an individual cancer patient is the key strategy to achieve a clinically effective treatment (273). Personalized investigation in cancer therapy includes genomic DNA sequencing, genomic exons sequencing, cancer-specific mutant genes sequencing, dysfunction of signaling proteins analysis, neoantigen detection, loss of immune cytotoxicity, and diversity of the TME (274). However, following nearly one century training, oncological clinicians have been used to dogmatically applying conventional chemotherapy and radiotherapy in almost all cancer treatment. Whether therapy-induced immune alterations are responsible for an effective therapy is less attractive in main menu despite the critical role of cellular immunotherapy to malignancy having been successfully exhibited through Dr Coley’s vaccine in 1893 (275). Actually, Coley’s vaccine strongly indicates that turning cold tumor to hot through immune modulation is an essential conversion from treatable to clinically curable. Currently, cellular immunotherapy includes neoantigens and vaccines, adoptive cell therapy, CAR-T/NK techniques, and oncolytic virus (OV) in purpose to recover immune normalization, reduce immunosuppression in the TME, increase tumor-specific antigen expression, and locally rebuild inflammatory conditions, as well as rearrange tumor vasculature.
Combination of Conventional Chemotherapy and Radiotherapy With Cellular Immunotherapy
For many years, a number of investigations have demonstrated that using pan-cytotoxic chemotherapy in cancer treatment causes a direct immunosuppression (276, 277), PDL-1 overexpression (278), recruitment of CSC-mediated drug resistance (200), and increased VEGFR-1-activated metastases (279), as well as myelosuppression (280), all of which objectively and comprehensively indicate that conventional chemotherapy is not an approach of precise medicine in cancer treatment. Surprisingly, through combining with certain kinds of chemotherapeutic agents, cellular immunotherapy significantly increases overall survival than singly using chemotherapy (281, 282). Further analysis elucidates that chemotherapeutics specifically acting on MDSCs and CSCs to reduce immunosuppression in the TME remarkably amplify efficiency and efficacy of ICI treatment and adoptive cell therapy (282, 283). Previously, clinical professionals supposed that apoptotic cancer cells or chemotherapy-induced mutation should have expressed more neoantigens; however, this idea is highly likely a misconception (284), but through applying histone deacetylase inhibitors or radioactive treatments, neoantigens expressed on cancer cells are notably beneficial for cellular immunotherapy due to enormously increased antigenicity (285, 286). Moreover, boron-neutron capture therapy (BNCT) not only precisely and largely eliminates tumor volume but also produces a large amount of various neoantigens due to radiation-damaged DNAs extensively existing in survived cancer cells, which are specifically utilized by cellular immunotherapy for constantly attacking (287, 288).
Adoptive Cell Therapy
Adoptive cell therapy includes TILs, cytokine-induced killer cells (CIK), dendritic cell-activated cytokine-induced killer cells (DC-CIK), NK cells, CAR-NK (chimeric antigen receptor-natural killer cells), and CAR-T (chimeric antigen receptor-T cells), in which TIL methodology showed clinical effects firstly on patients with metastatic melanoma in 1983 (289), and currently, TIL (290), DC-CIK (291), NK (292), CAR-NK (293), and CAR-T (294) all have achieved convincing results in cancer treatment, especially through DC-CIK, NK, and CAR-NK approaches.
As given knowledge, DCs are the most powerful antigen presentation immunocytes, which directly command cytotoxic T lymphocytes to attack cancer cells; therefore, depending on DC-mediated recruitment of CIKs through tumor-associated antigens, DC-CIK therapy efficiently and specifically eliminates malignant cells (295). Through three meta-analysis studies of more than 8 thousand patients with gastric cancer and CRC, comparing to individual application of chemotherapy, CIK/DC-CIK plus chemotherapy significantly increases overall survival and progression-free survival in drug-resistant patients, and swiftly improves most adverse events caused by chemotherapy (28, 296, 297). Similar results are also exhibited in CIK/DC-CIK therapy of pancreatic cancer and HCC, especially in advanced-stage patients (298, 299). Interestingly, infusion of allogeneic CIKs has also demonstrated encouraging effects on hematological malignancies (300). Comparing to DC-CIK therapy, as innate immune system, NK cell treatment occupies a unique advantage, in which tumor-associated antigen presentation is not compulsory for NK cell cytotoxicity, cytokine release syndrome is less possible to occur, and human leukocyte antigen matching is not stringently required for donor NK cell infusion; therefore, despite autologous NK cells showing very limited clinical efficacy (301), allogeneic NK cells including semi-allogeneic NK cells, NK cells isolated from umbilical cord blood, and iPSC-derived NK cells all show extraordinary immune killing specificity to malignant tumors without graft-versus-host disease (53, 302). Multiple infusions of allogeneic NK cells to stage-III/IV pancreatic cancer patients significantly increase overall survival and progression-free survival to 13.6 months and 9.9 months, respectively (303), whereas to unresectable primary HCC, are 23.2 months and 15.1 months, respectively (304), both of which clinically identify allogeneic NK cell therapy as a novel and promising methodology in treatment of GI cancers. Recent investigations discovered that PD-1 and PDL-1 expressed on DCs and NK cells reduced DC-activated maturation of cytotoxic T lymphocytes (305, 306) and NK-mediated antigen presentation of DCs (307–312), but through applying ICIs, both DC-CIK and NK therapies exhibit the higher level of cytotoxic activity to cancer cells (313–315).
Through studying the principle of TILs, clinical scientists naively consider that arming T cells from cancer patients with Chimeric tumor-specific Antigen Receptors (CAR-T) is efficient to destroy cancer cells precisely, and CAR-T technique indeed achieves the success in treatment of some kinds of hematological malignancies (316). However, different from attacking an individual malignant cell with unique antigen expression in the bloodstream or in the lymphatic nodes, CAR-T therapy to solid tumors is like using the sharpness of a knife to fight with the thickness of a tank, disadvantages of which include immunosuppression of the TME in solid tumors always primarily focusing on T cells, loss of tumor-associated antigens at the surface of mature solid tumors, and high cell density of inside solid tumors stopping CAR-Ts entering into the bulk, as well as extremely lethal side effects caused by immunotoxicity to the normal cells co-expressing tumor-associated antigens (317, 318). Also, due to continuously selective pressure caused by specific CAR, cancer cells may highly possibly abandon unique antigen expression through endocytosis. Therefore, in purpose of promoting immune normalization but not enhancement (319), rebuilding normal immunity in cancer patients through cellular immunotherapy is far more essential than CAR expression. Due to specific cytotoxicity to malignant tumors and much less possibility of inducing cytokine storm, clinical scientists design CAR-NK technique through equipping NK cells with chimeric antigen receptors, and have applied CAR-NK in patients with relapsed and refractory acute myeloid leukemia to test safety and tolerance (320). Whether CAR-NK therapy has more therapeutical advantages in solid tumors than combinative treatment of DC-CIK plus allogeneic NK cells, clinicians still have a long way to go. Following identification of specific biomarkers of TAMs, MDSCs, CAFs and CSCs, CAR-NK therapy certainly makes a great contribution in breaking through immunosuppression in the TME and strongly provide a beneficiary immune infrastructure for other kinds of cancer therapies.
Vaccine and Oncolytic Virus
Vaccines used in cancer therapy mainly depend on transferring tumor-associated antigens to antigen presenting cells such as DCs, and then inducing CD4+ helper T cells and CD8+ cytotoxic T cells to eradicate cancer cells (321). Genetic vaccines including DNA and RNA vaccines are theoretically able to synthesize all sequences encoding targeted antigens; however, according to various levels of protein translation, immunogenicity produced by genetic vaccines is hard to control (322). Despite DNA vaccines achieving pathological regression in treatment of intraepithelial neoplasia caused by human papillomavirus infection (323), clinical trials have not shown effective results in solid tumors therapy such as breast cancer (324), CRC (325), prostate cancer (326), and melanoma (327). Similar to DNA vaccines, messenger RNA vaccines containing 20 epitopes inoculated for patients with advanced-stage GI cancers have not obtained clinical efficacy, although they are safe and have activated immune response to neoepitopes (328). Actually, DC vaccines are more appropriately applied for cancer therapy in that they are ex vivo induced through the whole cancer cells, tumor lysates, peptides, DNAs, and RANs, which are strongly functional on tumor-specific cytotoxic T cells to eliminate cancer cells. Therapeutic DC vaccines have been extensively used in treatment of GI cancers at phase II or III trials, in which DC vaccine combining with MAGE peptide safely promotes immune response to tumor-specific antigens and significantly reduces tumor marker expression in nearly 90% advanced GI cancer patients (329), but for accomplishment of more optimal prognosis, prior to using DC vaccines, it is critical to examine NK cell activity in the candidates (330). Furthermore, in situ vaccines generated through chemoradiotherapy show more beneficial to cancer patients than conventional vaccines in that there is no screening requisition of positive antigen expression (331, 332). Almost all clinical trials demonstrate that adding ICIs into vaccine therapy remarkably increase vaccine-mediated cytotoxicity, which partially explains why individually using vaccine therapy only achieves suboptimal impact in cancer treatment (333, 334). Currently, the challenges vaccine therapy needs to deal with are multiple immunogenic antigen expression, construction of highly potent vaccine vectors, and breakthrough of immunosuppression in the TME.
Oncolytic virotherapy includes various kinds of viruses targeting a large range of malignant tumors, in which cancer cells are vulnerable to viral infection due to absence of type I interferon system and mutations promoting attachment of viruses (335, 336). In cancer therapy, using adenoviruses as a platform to express tumor-specific antigens and tumor-suppressive proteins has exhibited the promising results in preliminary animal studies, and currently in human studies, telomelysin-targeted adenovirus is testing in treatment of esophageal cancer, whereas evaluation of ideal viral vectors is processing in gastric cancer therapy, as well as more clinical trials are manipulating in treatment of pancreatic cancer, primary HCC, and CRC (337). Currently, clinical scientists are also concentrated on myxoma virus due to absence of infection to human beings and extremely sensitive to cancer cells; however, there still have a distance for clinical application (338). Furthermore, in diagnosis of GI cancers, depending on specific expression of fluorescence and bioluminescence on infected cancer cells, OVs, being tracing signals, are used to accurately expose the border of malignant tissue in surgery, and precisely identify the primary tumor and metastatic microtumors through nuclear medical imaging techniques such as CT, MRI, SPECT, and PET-CT (339–343). Through practicing oncolytic virotherapy, some barriers clinicians have to quell include tumor heterogeneity, general immune elimination of OVs, and capability to hack into tumor bulk and induce more activated cytotoxic T cells in the TME.
Conclusion
To successfully apply personalized therapy in cancer treatment, it is crucial to set up an assessment of tumor heterogeneity (344). In this review, we seemly underrate the importance of tumoral heterogeneity, in fact, which has been broadly discussed in parts of diagnosis and cellular therapy. Due to high variability of single subclone- or multiple subclone-induced tumor evolution in the same tumor or metastatic regions of the same tumor, as well as in different types of GI cancers, intratumoral genomic heterogeneity is highly beneficial for early diagnosis via liquid biopsy (345–347), while intertumoral heterogeneity-mediated changes of temporal and spatial plasticity and stemness in the TME precisely support the significance of comprehensive therapeutic principle (348, 349). Complexity of tumor heterogeneity essentially reflects adaption of genomic heterogeneity to selective pressure and utility of angiogenesis and immunosuppression through the TME for distant metastasis (350–352). Tumor heterogeneity also strongly indicates that rebuilding immune normalization to block interaction of intertumoral heterogeneity with the TME is a practical methodology to alter cancer therapy to chronic disease management.
Through systematically discussing key contributions of various components in the TME to tumorigenesis and metastases, reliability of early diagnostic methodologies, and personalized application of cellular immunotherapy in GI malignancies, it has become more apparent that immunosuppression induced by TAMs, MDSCs, CAFs, and CSCs in the TME and autocrine-paracrine network supported by stromal cells and cytokines and growth factors, especially TGFβ, are critically responsible for resistance to anticancer therapy and significant reduction of overall survival. Therefore, based on personalized investigation of each cancer patient including seeking mutant target genes and neoantigens, evaluating immune cytotoxicity in the TME, and recovering immune normalization, precise cellular immunotherapy is highly likely to change cancer treatment into a state of managing chronic disorders and eventually achieve the coexistence via normalization of immune surveillance. Due to complex of the TME, precise methodologies used to detect GI cancers at early stage primarily depend on the personalized combination of M2PK, miRNAs, ctDNAs, cancer-produced exosomes, and CTCs with conventional tumor markers and serologically biochemical examinations, in which CTC methodology is far more beneficial in evaluation of treatment efficacy and prognosis. Prior to treatment, a solid tumor is like an onion, in which anticancer mechanism of each layer is highly changeable, thereby, one step-by-step comprehensive treatment plan primarily includes that using BNCT technique efficiently reduces tumor volume and tears the tumor shell, and then applying OVs targeting TAMs, MDSCs, and CSCs with ICIs or dual recognition antibody of TGFβ and PD-1/PDL-1 maximally destroys immunosuppression and amplifies immunotoxicity of infiltrating T cells in the TME; furthermore, alternative infusion of DC-CIKs and allogeneic NK cells significantly eliminates most cancer cells with or without neoantigen expression. Such a therapy plan remarkably shrinks and eradicates the whole onion layer by layer. For CTCs or temporary formation of metastatic microtumors, CAR-T and CAR-NK therapies predominantly contribute to remove them. Surely, through recovering immune normalization in cancer patients and high-risk population, prevention of cancers in the community is far more critical than diagnosis and treatment. Whether precisely selecting the personalized cellular immunotherapy based on the diversity of the TME or rigidly relying on the unspecific chemotherapy, your choice.
Author Contributions
HZ made outline, wrote and revised the draft, and finalized the review. XL made outline. All authors contributed to the article and approved the submitted version.
Conflict of Interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Publisher’s Note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
Abbreviation
GI, gastrointestinal; TME, tumor microenvironment; DCs, dendrite cells; NKs, natural killer cells; TAMs, tumor-associated macrophage cells; CAFs, cancer-associated fibroblast cells; MDSCs, myeloid-derived suppressor cells; TGFβ, transform growth factor β; PD-1, programmed cell death protein 1; PD-L1, program death protein ligand 1; ICIs, immune checkpoint inhibitors; TIL, tumor-infiltrating lymphocytes; IL2, interleukin-2; IFNγ, interferon gamma; Tregs, T regulatory cells; CTLA4, cytotoxic T-lymphocyte antigen 4; CSCs, cancer stem cells; EMT, epithelial-mesenchymal transition; ECM, extracellular matrix; MAPK, mitogen-activated protein kinase; PI3K-Akt, phosphatidylinositol 3 kinase-Akt; STAT3, Signal Transducer and Activator of Transcription 3; EAC, Esophageal adenocarcinoma; ESCC, Esophageal squamous cell carcinoma; Hp, H. pylori; EBV, Epstein Barr Virus; HCC, hepatocellular carcinoma; PDAC, pancreatic duct adenocarcinoma; CRC, colorectal cancer; M2PK, M2 pyruvate kinase; FOBT, fecal occult blood test; CTCs, Circulating tumor cells; AUC, area under curve; ctDNAs, circulating tumor DNAs; miRNAs, microRNAs; circRNA, circular RNAs; BNCT, boron-neutron capture therapy; CIK, cytokine-induced killer cells; DC-CIK, dendritic cell-activated cytokine-induced killer cells; CAR-NK, chimeric antigen receptor-natural killer cells; and CAR-T, chimeric antigen receptor-T cells; OV, oncolytic virus.
References
1. Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, et al. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology (2020) 159(1):335–49 e15. doi: 10.1053/j.gastro.2020.02.068
2. Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, et al. Role of Tumor Microenvironment in Tumorigenesis. J Cancer (2017) 8(5):761–73. doi: 10.7150/jca.17648
3. Martins D, Schmitt F. Microenvironment in Breast Tumorigenesis: Friend or Foe? Histol Histopathol (2019) 34(1):13–24. doi: 10.14670/HH-18-021
4. Xian W, McKeon F. Microenvironment Meets Lineage Complexity in Junctional Tumorigenesis. Nat Commun (2019) 10(1):3829. doi: 10.1038/s41467-019-11651-6
5. Tong Q, Wang XL, Li SB, Yang GL, Jin S, Gao ZY, et al. Combined Detection of IL-6 and IL-8 is Beneficial to the Diagnosis of Early Stage Esophageal Squamous Cell Cancer: A Preliminary Study Based on the Screening of Serum Markers Using Protein Chips. Onco Targets Ther (2018) 11:5777–87. doi: 10.2147/OTT.S171242
6. Schneider J, Schulze G. Comparison of Tumor M2-pyruvate Kinase (Tumor M2-PK), Carcinoembryonic Antigen (CEA), Carbohydrate Antigens CA 19-9 and CA 72-4 in the Diagnosis of Gastrointestinal Cancer. Anticancer Res (2003) 23(6D):5089–93.
7. Li D, Wang G, Mei X. Diagnosis of Cancer at Early Stages Based on the Multiplex Detection of Tumor Markers Using Metal Nanoclusters. Analyst (2020) 145(22):7150–61. doi: 10.1039/d0an01538e
8. Killock D. Diagnosis: CancerSEEK and Destroy - a Blood Test for Early Cancer Detection. Nat Rev Clin Oncol (2018) 15(3):133. doi: 10.1038/nrclinonc.2018.21
9. Chen X, Gole J, Gore A, He Q, Lu M, Min J, et al. Non-Invasive Early Detection of Cancer Four Years Before Conventional Diagnosis Using a Blood Test. Nat Commun (2020) 11(1):3475. doi: 10.1038/s41467-020-17316-z
10. Crosby D, Lyons N, Greenwood E, Harrison S, Hiom S, Moffat J, et al. A Roadmap for the Early Detection and Diagnosis of Cancer. Lancet Oncol (2020) 21(11):1397–9. doi: 10.1016/S1470-2045(20)30593-3
11. Schulze G. The Tumor Marker Tumor M2-PK: An Application in the Diagnosis of Gastrointestinal Cancer. Anticancer Res (2000) 20(6D):4961–4.
12. Thakkar S, Sharma D, Kalia K, Tekade RK. Tumor Microenvironment Targeted Nanotherapeutics for Cancer Therapy and Diagnosis: A Review. Acta Biomater (2020) 101:43–68. doi: 10.1016/j.actbio.2019.09.009
13. Dudley ME, Rosenberg SA. Adoptive-Cell-Transfer Therapy for the Treatment of Patients With Cancer. Nat Rev Cancer (2003) 3(9):666–75. doi: 10.1038/nrc1167
14. Marcus A, Eshhar Z. Allogeneic Adoptive Cell Transfer Therapy as a Potent Universal Treatment for Cancer. Oncotarget (2011) 2(7):525–6. doi: 10.18632/oncotarget.300
15. Villadangos JA. Antigen-Specific Impairment of Adoptive T-cell Therapy Against Cancer: Players, Mechanisms, Solutions and a Hypothesis. Immunol Rev (2016) 272(1):169–82. doi: 10.1111/imr.12433
16. Hinrichs CS, Rosenberg SA. Exploiting the Curative Potential of Adoptive T-cell Therapy for Cancer. Immunol Rev (2014) 257(1):56–71. doi: 10.1111/imr.12132
17. Burchett R, Walsh S, Wan Y, Bramson JL. A Rational Relationship: Oncolytic Virus Vaccines as Functional Partners for Adoptive T Cell Therapy. Cytokine Growth Factor Rev (2020) 56:149–59. doi: 10.1016/j.cytogfr.2020.07.003
18. Chau I. Clinical Development of PD-1/PD-L1 Immunotherapy for Gastrointestinal Cancers: Facts and Hopes. Clin Cancer Res (2017) 23(20):6002–11. doi: 10.1158/1078-0432.CCR-17-0020
19. Wang D, Lin J, Yang X, Long J, Bai Y, Yang X, et al. Combination Regimens With PD-1/PD-L1 Immune Checkpoint Inhibitors for Gastrointestinal Malignancies. J Hematol Oncol (2019) 12(1):42. doi: 10.1186/s13045-019-0730-9
20. Park H, Chen B, Ciorba MA. Progress in PD-1-based Immunotherapy: New Mechanistic Insight may Provide Expanded Hope for Application to Colon and Gastrointestinal Cancers. Gastroenterology (2017) 153(4):1162–3. doi: 10.1053/j.gastro.2017.08.050
21. Wang S, Li Y, Xing C, Ding C, Zhang H, Chen L, et al. Tumor Microenvironment in Chemoresistance, Metastasis and Immunotherapy of Pancreatic Cancer. Am J Cancer Res (2020) 10(7):1937–53.
22. Zahn LM. Effects of the Tumor Microenvironment. Science (2017) 355(6332):1386–8. doi: 10.1126/science.355.6332.1386-l
23. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, et al. Differential Antigen Processing by Dendritic Cell Subsets In Vivo. Science (2007) 315(5808):107–11. doi: 10.1126/science.1136080
24. Steinman RM, Kaplan G, Witmer MD, Cohn ZA. Identification of a Novel Cell Type in Peripheral Lymphoid Organs of Mice. V. Purification of Spleen Dendritic Cells, New Surface Markers, and Maintenance In Vitro. J Exp Med (1979) 149(1):1–16. doi: 10.1084/jem.149.1.1
25. Steinman RM, Pack M, Inaba K. Dendritic Cells in the T-cell Areas of Lymphoid Organs. Immunol Rev (1997) 156:25–37. doi: 10.1111/j.1600-065x.1997.tb00956.x
26. Gabrilovich D. Mechanisms and Functional Significance of Tumour-Induced Dendritic-Cell Defects. Nat Rev Immunol (2004) 4(12):941–52. doi: 10.1038/nri1498
27. Satpathy AT, Kc W, Albring JC, Edelson BT, Kretzer NM, Bhattacharya D, et al. Zbtb46 Expression Distinguishes Classical Dendritic Cells and Their Committed Progenitors From Other Immune Lineages. J Exp Med (2012) 209(6):1135–52. doi: 10.1084/jem.20120030
28. Du H, Yang J, Zhang Y. Cytokine-Induced Killer Cell/Dendritic Cell Combined With Cytokine-Induced Killer Cell Immunotherapy for Treating Advanced Gastrointestinal Cancer. BMC Cancer (2020) 20(1):357. doi: 10.1186/s12885-020-06860-y
29. Cai L, Zhang C, Wu J, Zhou W, Chen T. Decreased PD-1 Expression on Circulating CD4(+)T Cell and PD-L1 Expression on Myeloid Dendritic Cell Correlate With Clinical Manifestations in Systemic Juvenile Idiopathic Arthritis. Joint Bone Spine (2019) 86(1):61–8. doi: 10.1016/j.jbspin.2018.03.003
30. Jeong Y, Kim GB, Ji Y, Kwak GJ, Nam GH, Hong Y, et al. Dendritic Cell Activation by an E. Coli-Derived Monophosphoryl Lipid A Enhances the Efficacy of PD-1 Blockade. Cancer Lett (2020) 472:19–28. doi: 10.1016/j.canlet.2019.12.012
31. Lurje I, Hammerich L, Tacke F. Dendritic Cell and T Cell Crosstalk in Liver Fibrogenesis and Hepatocarcinogenesis: Implications for Prevention and Therapy of Liver Cancer. Int J Mol Sci (2020) 21(19):7378. doi: 10.3390/ijms21197378
32. Aloysius MM, Takhar A, Robins A, Eremin O. Dendritic Cell Biology, Dysfunction and Immunotherapy in Gastrointestinal Cancers. Surgeon (2006) 4(4):195–210. doi: 10.1016/s1479-666x(06)80061-2
33. Zhu S, Yang N, Wu J, Wang X, Wang W, Liu YJ, et al. Tumor Microenvironment-Related Dendritic Cell Deficiency: A Target to Enhance Tumor Immunotherapy. Pharmacol Res (2020) 159:104980. doi: 10.1016/j.phrs.2020.104980
34. Lin JH, Huffman AP, Wattenberg MM, Walter DM, Carpenter EL, Feldser DM, et al. Type 1 Conventional Dendritic Cells are Systemically Dysregulated Early in Pancreatic Carcinogenesis. J Exp Med (2020) 217(8):e20190673. doi: 10.1084/jem.20190673
35. Li W, Wang W, Liao P, Song K, Zhu Z, Wang K, et al. High Levels of Tumor-Infiltrating Lymphocytes Showed Better Clinical Outcomes in FOLFOX-treated Gastric Cancer Patients. Pharmacogenomics (2020) 21(11):751–9. doi: 10.2217/pgs-2019-0102
36. Okadome K, Baba Y, Yagi T, Kiyozumi Y, Ishimoto T, Iwatsuki M, et al. Prognostic Nutritional Index, Tumor-infiltrating Lymphocytes, and Prognosis in Patients With Esophageal Cancer. Ann Surg (2020) 271(4):693–700. doi: 10.1097/SLA.0000000000002985
37. Choi E, Chang MS, Byeon SJ, Jin H, Jung KC, Kim H, et al. Prognostic Perspectives of PD-L1 Combined With Tumor-Infiltrating Lymphocytes, Epstein-Barr Virus, and Microsatellite Instability in Gastric Carcinomas. Diagn Pathol (2020) 15(1):69. doi: 10.1186/s13000-020-00979-z
38. Jun SY, Park ES, Lee JJ, Chang HK, Jung ES, Oh YH, et al. Prognostic Significance of Stromal and Intraepithelial Tumor-Infiltrating Lymphocytes in Small Intestinal Adenocarcinoma. Am J Clin Pathol (2020) 153(1):105–18. doi: 10.1093/ajcp/aqz136
39. Idos GE, Kwok J, Bonthala N, Kysh L, Gruber SB, Qu C. The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis. Sci Rep (2020) 10(1):3360. doi: 10.1038/s41598-020-60255-4
40. Xiao B, Peng J, Wang Y, Deng Y, Ou Q, Wu X, et al. Prognostic Value of Tumor Infiltrating Lymphocytes Combined With PD-L1 Expression for Patients With Solitary Colorectal Cancer Liver Metastasis. Ann Transl Med (2020) 8(19):1221. doi: 10.21037/atm-20-2762a
41. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The Immune Contexture in Human Tumours: Impact on Clinical Outcome. Nat Rev Cancer (2012) 12(4):298–306. doi: 10.1038/nrc3245
42. Kawahata K, Misaki Y, Yamauchi M, Tsunekawa S, Setoguchi K, Miyazaki J, et al. Generation of CD4(+)CD25(+) Regulatory T Cells From Autoreactive T Cells Simultaneously With Their Negative Selection in the Thymus and From Nonautoreactive T Cells by Endogenous TCR Expression. J Immunol (2002) 168(9):4399–405. doi: 10.4049/jimmunol.168.9.4399
43. Raffin C, Vo LT, Bluestone JA. Treg Cell-Based Therapies: Challenges and Perspectives. Nat Rev Immunol (2020) 20(3):158–72. doi: 10.1038/s41577-019-0232-6
44. Hira SK, Rej A, Paladhi A, Singh R, Saha J, Mondal I, et al. Galunisertib Drives Treg Fragility and Promotes Dendritic Cell-Mediated Immunity Against Experimental Lymphoma. iScience (2020) 23(10):101623. doi: 10.1016/j.isci.2020.101623
45. Grygorowicz MA, Borycka IS, Nowak E, Paszkiewicz-Kozik E, Rymkiewicz G, Blachnio K, et al. Lenalidomide Potentiates CD4(+)CD25(+)Treg-related Suppression of Lymphoma B-cell Proliferation. Clin Exp Med (2017) 17(2):193–207. doi: 10.1007/s10238-016-0411-8
46. Zhu MX, Wan WL, Tian L, Hu K, Wang J, Wang YF, et al. [Prognostic Significance of Treg/Th17 Cells Imbalance in Patients With Diffuse Large B Cell Lymphoma]. Zhonghua Xue Ye Xue Za Zhi (2018) 39(6):507–10. doi: 10.3760/cma.j.issn.0253-2727.2018.06.014
47. Marquez-Medina D, Salla-Fortuny J, Salud-Salvia A. Role of Gamma-Delta T-cells in Cancer: Another Opening Door to Immunotherapy. Clin Transl Oncol (2012) 14(12):891–5. doi: 10.1007/s12094-012-0935-7
48. Alam SM, Clark JS, Leech V, Whitford P, George WD, Campbell AM. T Cell Receptor Gamma/Delta Expression on Lymphocyte Populations of Breast Cancer Patients. Immunol Lett (1992) 31(3):279–83. doi: 10.1016/0165-2478(92)90127-a
49. Raspollini MR, Castiglione F, Rossi Degl’innocenti D, Amunni G, Villanucci A, Garbini F, et al. Tumour-Infiltrating Gamma/Delta T-lymphocytes are Correlated With a Brief Disease-Free Interval in Advanced Ovarian Serous Carcinoma. Ann Oncol (2005) 16(4):590–6. doi: 10.1093/annonc/mdi112
50. Bouet-Toussaint F, Cabillic F, Toutirais O, Le Gallo M, Thomas de la Pintiere C, Daniel P, et al. Vgamma9Vdelta2 T Cell-Mediated Recognition of Human Solid Tumors. Potential for Immunotherapy of Hepatocellular and Colorectal Carcinomas. Cancer Immunol Immunother (2008) 57(4):531–9. doi: 10.1007/s00262-007-0391-3
51. Chitadze G, Oberg HH, Wesch D, Kabelitz D. The Ambiguous Role of Gammadelta T Lymphocytes in Antitumor Immunity. Trends Immunol (2017) 38(9):668–78. doi: 10.1016/j.it.2017.06.004
52. Fleming C, Morrissey S, Cai Y, Yan J. Gammadelta T Cells: Unexpected Regulators of Cancer Development and Progression. Trends Cancer (2017) 3(8):561–70. doi: 10.1016/j.trecan.2017.06.003
53. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of Natural Killer Cells. Nat Immunol (2008) 9(5):503–10. doi: 10.1038/ni1582
54. Polidoro MA, Mikulak J, Cazzetta V, Lleo A, Mavilio D, Torzilli G, et al. Tumor Microenvironment in Primary Liver Tumors: A Challenging Role of Natural Killer Cells. World J Gastroenterol (2020) 26(33):4900–18. doi: 10.3748/wjg.v26.i33.4900
55. Mugitani H, Suzuki M, Naruki Y, Ohtsuka S, Shinbo T. [Natural Killer Cell Activity in Gastrointestinal Cancer Patients–a Decline in Natural Killer Cell Activity in T Cells and the Action of Interferon on its Increase (Author’s Transl)]. Nihon Gan Chiryo Gakkai Shi (1981) 16(7):1371–6.
56. Sun C, Xu J, Huang Q, Huang M, Wen H, Zhang C, et al. High NKG2A Expression Contributes to NK Cell Exhaustion and Predicts a Poor Prognosis of Patients With Liver Cancer. Oncoimmunology (2017) 6(1):e1264562. doi: 10.1080/2162402X.2016.1264562
57. Lee YE, Ju A, Choi HW, Kim JC, Kim EE, Kim TS, et al. Rationally Designed Redirection of Natural Killer Cells Anchoring a Cytotoxic Ligand for Pancreatic Cancer Treatment. J Control Release (2020) 326:310–23. doi: 10.1016/j.jconrel.2020.07.016
58. Park DJ, Sung PS, Kim JH, Lee GW, Jang JW, Jung ES, et al. EpCAM-high Liver Cancer Stem Cells Resist Natural Killer Cell-Mediated Cytotoxicity by Upregulating CEACAM1. J Immunother Cancer (2020) 8(1):e000301. doi: 10.1136/jitc-2019-000301
59. Mosser DM, Edwards JP. Exploring the Full Spectrum of Macrophage Activation. Nat Rev Immunol (2008) 8(12):958–69. doi: 10.1038/nri2448
60. Mantovani A, Sica A, Allavena P, Garlanda C, Locati M. Tumor-Associated Macrophages and the Related Myeloid-Derived Suppressor Cells as a Paradigm of the Diversity of Macrophage Activation. Hum Immunol (2009) 70(5):325–30. doi: 10.1016/j.humimm.2009.02.008
61. Eisinger S, Sarhan D, Boura VF, Ibarlucea-Benitez I, Tyystjarvi S, Oliynyk G, et al. Targeting a Scavenger Receptor on Tumor-Associated Macrophages Activates Tumor Cell Killing by Natural Killer Cells. Proc Natl Acad Sci USA (2020) 117(50):32005–16. doi: 10.1073/pnas.2015343117
62. Ojalvo LS, Whittaker CA, Condeelis JS, Pollard JW. Gene Expression Analysis of Macrophages That Facilitate Tumor Invasion Supports a Role for Wnt-signaling in Mediating Their Activity in Primary Mammary Tumors. J Immunol (2010) 184(2):702–12. doi: 10.4049/jimmunol.0902360
63. D’Errico G, Alonso-Nocelo M, Vallespinos M, Hermann PC, Alcala S, Garcia CP, et al. Tumor-Associated Macrophage-Secreted 14-3-3zeta Signals Via AXL to Promote Pancreatic Cancer Chemoresistance. Oncogene (2019) 38(27):5469–85. doi: 10.1038/s41388-019-0803-9
64. Kurahara H, Shinchi H, Mataki Y, Maemura K, Noma H, Kubo F, et al. Significance of M2-polarized Tumor-Associated Macrophage in Pancreatic Cancer. J Surg Res (2011) 167(2):e211–9. doi: 10.1016/j.jss.2009.05.026
65. Zheng Y, Cai Z, Wang S, Zhang X, Qian J, Hong S, et al. Macrophages are an Abundant Component of Myeloma Microenvironment and Protect Myeloma Cells From Chemotherapy Drug-Induced Apoptosis. Blood (2009) 114(17):3625–8. doi: 10.1182/blood-2009-05-220285
66. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated Regulation of Myeloid Cells by Tumours. Nat Rev Immunol (2012) 12(4):253–68. doi: 10.1038/nri3175
67. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, et al. Expansion of Myeloid Immune Suppressor Gr+CD11b+ Cells in Tumor-Bearing Host Directly Promotes Tumor Angiogenesis. Cancer Cell (2004) 6(4):409–21. doi: 10.1016/j.ccr.2004.08.031
68. Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI. Mechanism of T Cell Tolerance Induced by Myeloid-Derived Suppressor Cells. J Immunol (2010) 184(6):3106–16. doi: 10.4049/jimmunol.0902661
69. Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, et al. Arginase I Production in the Tumor Microenvironment by Mature Myeloid Cells Inhibits T-cell Receptor Expression and Antigen-Specific T-cell Responses. Cancer Res (2004) 64(16):5839–49. doi: 10.1158/0008-5472.CAN-04-0465
70. Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-Derived Suppressor Cells Inhibit T-cell Activation by Depleting Cystine and Cysteine. Cancer Res (2010) 70(1):68–77. doi: 10.1158/0008-5472.CAN-09-2587
71. Molon B, Ugel S, Del Pozzo F, Soldani C, Zilio S, Avella D, et al. Chemokine Nitration Prevents Intratumoral Infiltration of Antigen-Specific T Cells. J Exp Med (2011) 208(10):1949–62. doi: 10.1084/jem.20101956
72. Pan PY, Ma G, Weber KJ, Ozao-Choy J, Wang G, Yin B, et al. Immune Stimulatory Receptor CD40 is Required for T-cell Suppression and T Regulatory Cell Activation Mediated by Myeloid-Derived Suppressor Cells in Cancer. Cancer Res (2010) 70(1):99–108. doi: 10.1158/0008-5472.CAN-09-1882
73. Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-Derived Suppressor Cells Promote Cross-Tolerance in B-cell Lymphoma by Expanding Regulatory T Cells. Cancer Res (2008) 68(13):5439–49. doi: 10.1158/0008-5472.CAN-07-6621
74. Cao S, Liu J, Chesi M, Bergsagel PL, Ho IC, Donnelly RP, et al. Differential Regulation of IL-12 and IL-10 Gene Expression in Macrophages by the Basic Leucine Zipper Transcription Factor c-Maf Fibrosarcoma. J Immunol (2002) 169(10):5715–25. doi: 10.4049/jimmunol.169.10.5715
75. Murai M, Turovskaya O, Kim G, Madan R, Karp CL, Cheroutre H, et al. Interleukin 10 Acts on Regulatory T Cells to Maintain Expression of the Transcription Factor Foxp3 and Suppressive Function in Mice With Colitis. Nat Immunol (2009) 10(11):1178–84. doi: 10.1038/ni.1791
76. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, et al. Cd4(+) T Cells Regulate Pulmonary Metastasis of Mammary Carcinomas by Enhancing Protumor Properties of Macrophages. Cancer Cell (2009) 16(2):91–102. doi: 10.1016/j.ccr.2009.06.018
77. Elkabets M, Ribeiro VS, Dinarello CA, Ostrand-Rosenberg S, Di Santo JP, Apte RN, et al. Il-1beta Regulates a Novel Myeloid-Derived Suppressor Cell Subset That Impairs NK Cell Development and Function. Eur J Immunol (2010) 40(12):3347–57. doi: 10.1002/eji.201041037
78. Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, Smith K, et al. Il-23 Promotes Tumour Incidence and Growth. Nature (2006) 442(7101):461–5. doi: 10.1038/nature04808
79. Fabian KP, Padget MR, Donahue RN, Solocinski K, Robbins Y, Allen CT, et al. Pd-L1 Targeting High-Affinity NK (t-haNK) Cells Induce Direct Antitumor Effects and Target Suppressive MDSC Populations. J Immunother Cancer (2020) 8(1):e000450. doi: 10.1136/jitc-2019-000450
80. Greene S, Robbins Y, Mydlarz WK, Huynh AP, Schmitt NC, Friedman J, et al. Inhibition of MDSC Trafficking With SX-682, a CXCR1/2 Inhibitor, Enhances Nk-Cell Immunotherapy in Head and Neck Cancer Models. Clin Cancer Res (2020) 26(6):1420–31. doi: 10.1158/1078-0432.CCR-19-2625
81. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. Il-23 Drives a Pathogenic T Cell Population That Induces Autoimmune Inflammation. J Exp Med (2005) 201(2):233–40. doi: 10.1084/jem.20041257
82. Poschke I, Mao Y, Adamson L, Salazar-Onfray F, Masucci G, Kiessling R. Myeloid-Derived Suppressor Cells Impair the Quality of Dendritic Cell Vaccines. Cancer Immunol Immunother (2012) 61(6):827–38. doi: 10.1007/s00262-011-1143-y
83. Zhang J, Li Z, Hu X, Su Q, He C, Liu J, et al. Knockout of P2Y12 Aggravates Experimental Autoimmune Encephalomyelitis in Mice Via Increasing of IL-23 Production and Th17 Cell Differentiation by Dendritic Cells. Brain Behav Immun (2017) 62:245–55. doi: 10.1016/j.bbi.2016.12.001
84. Li B, Wang JH. Fibroblasts and Myofibroblasts in Wound Healing: Force Generation and Measurement. J Tissue Viability (2011) 20(4):108–20. doi: 10.1016/j.jtv.2009.11.004
85. Rockey DC, Weymouth N, Shi Z. Smooth Muscle Alpha Actin (Acta2) and Myofibroblast Function During Hepatic Wound Healing. PloS One (2013) 8(10):e77166. doi: 10.1371/journal.pone.0077166
86. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of Fibroblast Heterogeneity in the Tumor Microenvironment. Cancer Biol Ther (2006) 5(12):1640–6. doi: 10.4161/cbt.5.12.3354
87. Dong R, Guo J, Zhang Z, Zhou Y, Hua Y. Polyphyllin I Inhibits Gastric Cancer Cell Proliferation by Downregulating the Expression of Fibroblast Activation Protein Alpha (FAP) and Hepatocyte Growth Factor (HGF) in Cancer-Associated Fibroblasts. Biochem Biophys Res Commun (2018) 497(4):1129–34. doi: 10.1016/j.bbrc.2018.02.193
88. Fukumura D, Xavier R, Sugiura T, Chen Y, Park EC, Lu N, et al. Tumor Induction of VEGF Promoter Activity in Stromal Cells. Cell (1998) 94(6):715–25. doi: 10.1016/s0092-8674(00)81731-6
89. Monteran L, Erez N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front Immunol (2019) 10:1835. doi: 10.3389/fimmu.2019.01835
90. Fearon DT. The Carcinoma-Associated Fibroblast Expressing Fibroblast Activation Protein and Escape From Immune Surveillance. Cancer Immunol Res (2014) 2(3):187–93. doi: 10.1158/2326-6066.Cir-14-0002
91. Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 From FAP-expressing Carcinoma-Associated Fibroblasts Synergizes With anti-PD-L1 Immunotherapy in Pancreatic Cancer. Proc Natl Acad Sci USA (2013) 110(50):20212–7. doi: 10.1073/pnas.1320318110
92. Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting Focal Adhesion Kinase Renders Pancreatic Cancers Responsive to Checkpoint Immunotherapy. Nat Med (2016) 22(8):851–60. doi: 10.1038/nm.4123
93. Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-Associated Fibroblasts are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell (2010) 17(2):135–47. doi: 10.1016/j.ccr.2009.12.041
94. Johansson AC, Ansell A, Jerhammar F, Lindh MB, Grenman R, Munck-Wikland E, et al. Cancer-Associated Fibroblasts Induce Matrix Metalloproteinase-Mediated Cetuximab Resistance in Head and Neck Squamous Cell Carcinoma Cells. Mol Cancer Res (2012) 10(9):1158–68. doi: 10.1158/1541-7786.MCR-12-0030
95. Yegodayev KM, Novoplansky O, Golden A, Prasad M, Levin L, Jagadeeshan S, et al. Tgf-Beta-Activated Cancer-Associated Fibroblasts Limit Cetuximab Efficacy in Preclinical Models of Head and Neck Cancer. Cancers (Basel) (2020) 12(2):339. doi: 10.3390/cancers12020339
96. Katuri V, Tang Y, Marshall B, Rashid A, Jogunoori W, Volpe EA, et al. Inactivation of ELF/TGF-beta Signaling in Human Gastrointestinal Cancer. Oncogene (2005) 24(54):8012–24. doi: 10.1038/sj.onc.1208946
97. Principe DR, DeCant B, Mascarinas E, Wayne EA, Diaz AM, Akagi N, et al. Tgfbeta Signaling in the Pancreatic Tumor Microenvironment Promotes Fibrosis and Immune Evasion to Facilitate Tumorigenesis. Cancer Res (2016) 76(9):2525–39. doi: 10.1158/0008-5472.CAN-15-1293
98. Lebrun JJ. The Dual Role of TGFbeta in Human Cancer: From Tumor Suppression to Cancer Metastasis. ISRN Mol Biol (2012) 2012:381428. doi: 10.5402/2012/381428
100. Massague J. Tgfbeta Signalling in Context. Nat Rev Mol Cell Biol (2012) 13(10):616–30. doi: 10.1038/nrm3434
101. Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL. p38 Mitogen-Activated Protein Kinase is Required for TGFbeta-mediated Fibroblastic Transdifferentiation and Cell Migration. J Cell Sci (2002) 115(Pt 15):3193–206. doi: 10.1242/jcs.115.15.3193
102. Rahimi RA, Leof EB. Tgfbeta Versatility: PI3K as a Critical Mediator of Distinct Cell Type and Context Specific Responses. Cell Cycle (2009) 8(12):1813–4. doi: 10.4161/cc.8.12.8828
103. Feng XH, Liang YY, Liang M, Zhai W, Lin X. Direct Interaction of c-Myc With Smad2 and Smad3 to Inhibit TGF-Beta-Mediated Induction of the CDK Inhibitor P15(Ink4B). Mol Cell (2016) 63(6):1089. doi: 10.1016/j.molcel.2016.08.027
104. Sanchez-Capelo A. Dual Role for TGF-beta1 in Apoptosis. Cytokine Growth Factor Rev (2005) 16(1):15–34. doi: 10.1016/j.cytogfr.2004.11.002
105. Cassar L, Li H, Jiang FX, Liu JP. TGF-Beta Induces Telomerase-Dependent Pancreatic Tumor Cell Cycle Arrest. Mol Cell Endocrinol (2010) 320(1-2):97–105. doi: 10.1016/j.mce.2010.02.002
106. Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, et al. Tgfbeta Drives Immune Evasion in Genetically Reconstituted Colon Cancer Metastasis. Nature (2018) 554(7693):538–43. doi: 10.1038/nature25492
107. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. Tgfbeta Attenuates Tumour Response to PD-L1 Blockade by Contributing to Exclusion of T Cells. Nature (2018) 554(7693):544–8. doi: 10.1038/nature25501
108. Huang CY, Chung CL, Hu TH, Chen JJ, Liu PF, Chen CL. Recent Progress in TGF-beta Inhibitors for Cancer Therapy. BioMed Pharmacother (2021) 134:111046. doi: 10.1016/j.biopha.2020.111046
109. Lind H, Gameiro SR, Jochems C, Donahue RN, Strauss J, Gulley JM, et al. Dual Targeting of TGF-beta and PD-L1 Via a Bifunctional anti-PD-L1/TGF-betaRII Agent: Status of Preclinical and Clinical Advances. J Immunother Cancer (2020) 8(1):e000433. doi: 10.1136/jitc-2019-000433
110. Coussens LM, Werb Z. Inflammation and Cancer. Nature (2002) 420(6917):860–7. doi: 10.1038/nature01322
111. Dvorakova K, Payne CM, Ramsey L, Holubec H, Sampliner R, Dominguez J, et al. Increased Expression and Secretion of Interleukin-6 in Patients With Barrett’s Esophagus. Clin Cancer Res (2004) 10(6):2020–8. doi: 10.1158/1078-0432.ccr-0437-03
112. Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, et al. Regulation of the Innate and Adaptive Immune Responses by Stat-3 Signaling in Tumor Cells. Nat Med (2004) 10(1):48–54. doi: 10.1038/nm976
113. Huang FL, Yu SJ. Esophageal Cancer: Risk Factors, Genetic Association, and Treatment. Asian J Surg (2018) 41(3):210–5. doi: 10.1016/j.asjsur.2016.10.005
114. Tanaka Y, Yoshida K, Suetsugu T, Imai T, Matsuhashi N, Yamaguchi K. Recent Advancements in Esophageal Cancer Treatment in Japan. Ann Gastroenterol Surg (2018) 2(4):253–65. doi: 10.1002/ags3.12174
115. Karakasheva TA, Waldron TJ, Eruslanov E, Kim SB, Lee JS, O’Brien S, et al. Cd38-Expressing Myeloid-Derived Suppressor Cells Promote Tumor Growth in a Murine Model of Esophageal Cancer. Cancer Res (2015) 75(19):4074–85. doi: 10.1158/0008-5472.CAN-14-3639
116. Pal S, Nandi M, Dey D, Chakraborty BC, Shil A, Ghosh S, et al. Myeloid-Derived Suppressor Cells Induce Regulatory T Cells in Chronically HBV Infected Patients With High Levels of Hepatitis B Surface Antigen and Persist After Antiviral Therapy. Aliment Pharmacol Ther (2019) 49(10):1346–59. doi: 10.1111/apt.15226
117. Sohda M, Kuwano H. Current Status and Future Prospects for Esophageal Cancer Treatment. Ann Thorac Cardiovasc Surg (2017) 23(1):1–11. doi: 10.5761/atcs.ra.16-00162
118. Miyashita T, Tajima H, Shah FA, Oshima M, Makino I, Nakagawara H, et al. Impact of Inflammation-Metaplasia-Adenocarcinoma Sequence and Inflammatory Microenvironment in Esophageal Carcinogenesis Using Surgical Rat Models. Ann Surg Oncol (2014) 21(6):2012–9. doi: 10.1245/s10434-014-3537-5
119. Wang J, Zhang G, Wang J, Wang L, Huang X, Cheng Y. The Role of Cancer-Associated Fibroblasts in Esophageal Cancer. J Transl Med (2016) 14:30. doi: 10.1186/s12967-016-0788-x
120. Tomas-Bort E, Kieler M, Sharma S, Candido JB, Loessner D. 3D Approaches to Model the Tumor Microenvironment of Pancreatic Cancer. Theranostics (2020) 10(11):5074–89. doi: 10.7150/thno.42441
121. Polk DB, Peek RM Jr. Helicobacter Pylori: Gastric Cancer and Beyond. Nat Rev Cancer (2010) 10(6):403–14. doi: 10.1038/nrc2857
122. Rasch S, Algul H. A Clinical Perspective on the Role of Chronic Inflammation in Gastrointestinal Cancer. Clin Exp Gastroenterol (2014) 7:261–72. doi: 10.2147/CEG.S43457
123. Lee HJ, Park JM, Han YM, Gil HK, Kim J, Chang JY, et al. The Role of Chronic Inflammation in the Development of Gastrointestinal Cancers: Reviewing Cancer Prevention With Natural Anti-Inflammatory Intervention. Expert Rev Gastroenterol Hepatol (2016) 10(1):129–39. doi: 10.1586/17474124.2016.1103179
125. Gyori D, Chessa T, Hawkins PT, Stephens LR. Class (I) Phosphoinositide 3-Kinases in the Tumor Microenvironment. Cancers (Basel) (2017) 9(3):24. doi: 10.3390/cancers9030024
126. Zhou H, Liu H, Jiang M, Zhang S, Chen J, Fan X. Targeting MicroRNA-21 Suppresses Gastric Cancer Cell Proliferation and Migration Via PTEN/Akt Signaling Axis. Cell Transplant (2019) 28(3):306–17. doi: 10.1177/0963689719825573
127. Wang Z, Ye Y, Liu D, Yang X, Wang F. Hypermethylation of Multiple Wnt Antagonist Genes in Gastric Neoplasia: Is H Pylori Infection Blasting Fuse? Med (Baltimore) (2018) 97(52):e13734. doi: 10.1097/MD.0000000000013734
128. Murata-Kamiya N, Kurashima Y, Teishikata Y, Yamahashi Y, Saito Y, Higashi H, et al. Helicobacter Pylori CagA Interacts With E-cadherin and Deregulates the Beta-Catenin Signal That Promotes Intestinal Transdifferentiation in Gastric Epithelial Cells. Oncogene (2007) 26(32):4617–26. doi: 10.1038/sj.onc.1210251
129. Fujii Y, Yoshihashi K, Suzuki H, Tsutsumi S, Mutoh H, Maeda S, et al. CDX1 Confers Intestinal Phenotype on Gastric Epithelial Cells Via Induction of Stemness-Associated Reprogramming Factors SALL4 and KLF5. Proc Natl Acad Sci USA (2012) 109(50):20584–9. doi: 10.1073/pnas.1208651109
130. Zhang L, Xu Z, Xu X, Zhang B, Wu H, Wang M, et al. SALL4, a Novel Marker for Human Gastric Carcinogenesis and Metastasis. Oncogene (2014) 33(48):5491–500. doi: 10.1038/onc.2013.495
131. Choi YJ, Kim N, Chang H, Lee HS, Park SM, Park JH, et al. Helicobacter Pylori-Induced Epithelial-Mesenchymal Transition, a Potential Role of Gastric Cancer Initiation and an Emergence of Stem Cells. Carcinogenesis (2015) 36(5):553–63. doi: 10.1093/carcin/bgv022
132. Terashima M, Kitada K, Ochiai A, Ichikawa W, Kurahashi I, Sakuramoto S, et al. Impact of Expression of Human Epidermal Growth Factor Receptors EGFR and ERBB2 on Survival in Stage II/III Gastric Cancer. Clin Cancer Res (2012) 18(21):5992–6000. doi: 10.1158/1078-0432.CCR-12-1318
133. Xu W, Yang Z, Lu N. Molecular Targeted Therapy for the Treatment of Gastric Cancer. J Exp Clin Cancer Res (2016) 35:1. doi: 10.1186/s13046-015-0276-9
134. Zhang Z, Li Z, Li Y, Zang A. MicroRNA and Signaling Pathways in Gastric Cancer. Cancer Gene Ther (2014) 21(8):305–16. doi: 10.1038/cgt.2014.37
135. Chen X, Gao H, Deng Y, Jin Q, Ji J, Ding D. Supramolecular Aggregation-Induced Emission Nanodots With Programmed Tumor Microenvironment Responsiveness for Image-Guided Orthotopic Pancreatic Cancer Therapy. ACS Nano (2020) 14(4):5121–34. doi: 10.1021/acsnano.0c02197
136. Ma L, Hernandez MO, Zhao Y, Mehta M, Tran B, Kelly M, et al. Tumor Cell Biodiversity Drives Microenvironmental Reprogramming in Liver Cancer. Cancer Cell (2019) 36(4):418–30 e6. doi: 10.1016/j.ccell.2019.08.007
137. Shen YC, Hsu C, Cheng AL. Molecular Targeted Therapy for Advanced Hepatocellular Carcinoma: Current Status and Future Perspectives. J Gastroenterol (2010) 45(8):794–807. doi: 10.1007/s00535-010-0270-0
138. Yoo YG, Oh SH, Park ES, Cho H, Lee N, Park H, et al. Hepatitis B Virus X Protein Enhances Transcriptional Activity of Hypoxia-Inducible factor-1alpha Through Activation of Mitogen-Activated Protein Kinase Pathway. J Biol Chem (2003) 278(40):39076–84. doi: 10.1074/jbc.M305101200
139. Thomas H. Liver Cancer: Lenvatinib non-Inferior to Sorafenib for Hepatocellular Carcinoma. Nat Rev Gastroenterol Hepatol (2018) 15(4):190. doi: 10.1038/nrgastro.2018.20
140. Oh SH, Swiderska-Syn M, Jewell ML, Premont RT, Diehl AM. Liver Regeneration Requires Yap1-TGFbeta-dependent Epithelial-Mesenchymal Transition in Hepatocytes. J Hepatol (2018) 69(2):359–67. doi: 10.1016/j.jhep.2018.05.008
141. Zhou SJ, Liu FY, Jiang YH, Liang HF. Reply to ‘Comment on ‘MicroRNA-199b-5p Attenuates TGF-beta1-induced Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma’’. Br J Cancer (2018) 118(7):1030. doi: 10.1038/s41416-018-0031-z
142. Bi JG, Zheng JF, Li Q, Bao SY, Yu XF, Xu P, et al. MicroRNA-181a-5p Suppresses Cell Proliferation by Targeting Egr1 and Inhibiting Egr1/TGF-beta/Smad Pathway in Hepatocellular Carcinoma. Int J Biochem Cell Biol (2019) 106:107–16. doi: 10.1016/j.biocel.2018.11.011
143. Zhou Q, Lui VW, Yeo W. Targeting the PI3K/Akt/mTOR Pathway in Hepatocellular Carcinoma. Future Oncol (2011) 7(10):1149–67. doi: 10.2217/fon.11.95
144. Hu TH, Huang CC, Lin PR, Chang HW, Ger LP, Lin YW, et al. Expression and Prognostic Role of Tumor Suppressor Gene PTEN/MMAC1/TEP1 in Hepatocellular Carcinoma. Cancer (2003) 97(8):1929–40. doi: 10.1002/cncr.11266
145. Song XZ, Ren XN, Xu XJ, Ruan XX, Wang YL, Yao TT. Lncrna RHPN1-AS1 Promotes Cell Proliferation, Migration and Invasion Through Targeting miR-7-5p and Activating PI3K/AKT/Mtor Pathway in Hepatocellular Carcinoma. Technol Cancer Res Treat (2020) 19:1533033820957023. doi: 10.1177/1533033820957023
146. Zhu AX, Abrams TA, Miksad R, Blaszkowsky LS, Meyerhardt JA, Zheng H, et al. Phase 1/2 Study of Everolimus in Advanced Hepatocellular Carcinoma. Cancer (2011) 117(22):5094–102. doi: 10.1002/cncr.26165
147. Kirstein MM, Boukouris AE, Pothiraju D, Buitrago-Molina LE, Marhenke S, Schutt J, et al. Activity of the mTOR Inhibitor RAD001, the Dual mTOR and PI3-kinase Inhibitor BEZ235 and the PI3-kinase Inhibitor BKM120 in Hepatocellular Carcinoma. Liver Int (2013) 33(5):780–93. doi: 10.1111/liv.12126
148. Wong CM, Fan ST, Ng IO. beta-Catenin Mutation and Overexpression in Hepatocellular Carcinoma: Clinicopathologic and Prognostic Significance. Cancer (2001) 92(1):136–45. doi: 10.1002/1097-0142(20010701)92:1<136::aid-cncr1301>3.0.co;2-r
149. Giles RH, van Es JH, Clevers H. Caught Up in a Wnt Storm: Wnt Signaling in Cancer. Biochim Biophys Acta (2003) 1653(1):1–24. doi: 10.1016/s0304-419x(03)00005-2
150. Hoshida Y, Nijman SM, Kobayashi M, Chan JA, Brunet JP, Chiang DY, et al. Integrative Transcriptome Analysis Reveals Common Molecular Subclasses of Human Hepatocellular Carcinoma. Cancer Res (2009) 69(18):7385–92. doi: 10.1158/0008-5472.CAN-09-1089
151. Tan J, Tong BD, Wu YJ, Xiong W. MicroRNA-29 Mediates TGFbeta1-induced Extracellular Matrix Synthesis by Targeting Wnt/Beta-Catenin Pathway in Human Orbital Fibroblasts. Int J Clin Exp Pathol (2014) 7(11):7571–7.
152. Zhang H, Ye J, Weng X, Liu F, He L, Zhou D, et al. Comparative Transcriptome Analysis Reveals That the Extracellular Matrix Receptor Interaction Contributes to the Venous Metastases of Hepatocellular Carcinoma. Cancer Genet (2015) 208(10):482–91. doi: 10.1016/j.cancergen.2015.06.002
153. Carloni V, Luong TV, Rombouts K. Hepatic Stellate Cells and Extracellular Matrix in Hepatocellular Carcinoma: More Complicated Than Ever. Liver Int (2014) 34(6):834–43. doi: 10.1111/liv.12465
154. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2020. CA Cancer J Clin (2020) 70(1):7–30. doi: 10.3322/caac.21590
155. Adamska A, Domenichini A, Falasca M. Pancreatic Ductal Adenocarcinoma: Current and Evolving Therapies. Int J Mol Sci (2017) 18(7):1338. doi: 10.3390/ijms18071338
156. Paniccia A, Hosokawa P, Henderson W, Schulick RD, Edil BH, McCarter MD, et al. Characteristics of 10-Year Survivors of Pancreatic Ductal Adenocarcinoma. JAMA Surg (2015) 150(8):701–10. doi: 10.1001/jamasurg.2015.0668
157. Nakayama H, Ohuchida K, Yoshida M, Miyazaki T, Takesue S, Abe T, et al. Degree of Desmoplasia in Metastatic Lymph Node Lesions is Associated With Lesion Size and Poor Prognosis in Pancreatic Cancer Patients. Oncol Lett (2017) 14(3):3141–7. doi: 10.3892/ol.2017.6549
158. Iglesias M, Frontelo P, Gamallo C, Quintanilla M. Blockade of Smad4 in Transformed Keratinocytes Containing a Ras Oncogene Leads to Hyperactivation of the Ras-dependent Erk Signalling Pathway Associated With Progression to Undifferentiated Carcinomas. Oncogene (2000) 19(36):4134–45. doi: 10.1038/sj.onc.1203764
159. Tan Z, Xu J, Zhang B, Shi S, Yu X, Liang C. Hypoxia: A Barricade to Conquer the Pancreatic Cancer. Cell Mol Life Sci (2020) 77(16):3077–83. doi: 10.1007/s00018-019-03444-3
160. Samkharadze T, Erkan M, Reiser-Erkan C, Demir IE, Kong B, Ceyhan GO, et al. Pigment Epithelium-Derived Factor Associates With Neuropathy and Fibrosis in Pancreatic Cancer. Am J Gastroenterol (2011) 106(5):968–80. doi: 10.1038/ajg.2010.479
161. Kisker O, Onizuka S, Banyard J, Komiyama T, Becker CM, Achilles EG, et al. Generation of Multiple Angiogenesis Inhibitors by Human Pancreatic Cancer. Cancer Res (2001) 61(19):7298–304.
162. Xue Y, Beyer G, Mayerle J. Fapalpha in Pancreatic Stellate Cells Upregulated by TGFbeta1: A Novel Insight Into Pancreatic Cancer Progress. Ann Transl Med (2020) 8(15):910. doi: 10.21037/atm-20-2559
163. Pothula SP, Pirola RC, Wilson JS, Apte MV. Pancreatic Stellate Cells: Aiding and Abetting Pancreatic Cancer Progression. Pancreatology (2020) 20(3):409–18. doi: 10.1016/j.pan.2020.01.003
164. Mace TA, Ameen Z, Collins A, Wojcik S, Mair M, Young GS, et al. Pancreatic Cancer-Associated Stellate Cells Promote Differentiation of Myeloid-Derived Suppressor Cells in a STAT3-Dependent Manner. Cancer Res (2013) 73(10):3007–18. doi: 10.1158/0008-5472.CAN-12-4601
165. Tang D, Yuan Z, Xue X, Lu Z, Zhang Y, Wang H, et al. High Expression of Galectin-1 in Pancreatic Stellate Cells Plays a Role in the Development and Maintenance of an Immunosuppressive Microenvironment in Pancreatic Cancer. Int J Cancer (2012) 130(10):2337–48. doi: 10.1002/ijc.26290
166. Zhang Y, Ware MB, Zaidi MY, Ruggieri AN, Olson BM, Komar H, et al. Heat Shock Protein-90 Inhibition Alters Activation of Pancreatic Stellate Cells and Enhances the Efficacy of PD-1 Blockade in Pancreatic Cancer. Mol Cancer Ther (2021) 20(1):150–60. doi: 10.1158/1535-7163.MCT-19-0911
167. Masamune A, Kikuta K, Watanabe T, Satoh K, Hirota M, Shimosegawa T. Hypoxia Stimulates Pancreatic Stellate Cells to Induce Fibrosis and Angiogenesis in Pancreatic Cancer. Am J Physiol Gastrointest Liver Physiol (2008) 295(4):G709–17. doi: 10.1152/ajpgi.90356.2008
168. Szablowska-Gadomska I, Zayat V, Buzanska L. Influence of Low Oxygen Tensions on Expression of Pluripotency Genes in Stem Cells. Acta Neurobiol Exp (Wars) (2011) 71(1):86–93.
169. Hamada S, Masamune A, Takikawa T, Suzuki N, Kikuta K, Hirota M, et al. Pancreatic Stellate Cells Enhance Stem Cell-Like Phenotypes in Pancreatic Cancer Cells. Biochem Biophys Res Commun (2012) 421(2):349–54. doi: 10.1016/j.bbrc.2012.04.014
170. Cave DD, Di Guida M, Costa V, Sevillano M, Ferrante L, Heeschen C, et al. TGF-Beta1 Secreted by Pancreatic Stellate Cells Promotes Stemness and Tumourigenicity in Pancreatic Cancer Cells Through L1CAM Downregulation. Oncogene (2020) 39(21):4271–85. doi: 10.1038/s41388-020-1289-1
171. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. Distinct Populations of Cancer Stem Cells Determine Tumor Growth and Metastatic Activity in Human Pancreatic Cancer. Cell Stem Cell (2007) 1(3):313–23. doi: 10.1016/j.stem.2007.06.002
172. Quint K, Tonigold M, Di Fazio P, Montalbano R, Lingelbach S, Ruckert F, et al. Pancreatic Cancer Cells Surviving Gemcitabine Treatment Express Markers of Stem Cell Differentiation and Epithelial-Mesenchymal Transition. Int J Oncol (2012) 41(6):2093–102. doi: 10.3892/ijo.2012.1648
173. Polireddy K, Chen Q. Cancer of the Pancreas: Molecular Pathways and Current Advancement in Treatment. J Cancer (2016) 7(11):1497–514. doi: 10.7150/jca.14922
174. Lonardo E, Hermann PC, Mueller MT, Huber S, Balic A, Miranda-Lorenzo I, et al. Nodal/Activin Signaling Drives Self-Renewal and Tumorigenicity of Pancreatic Cancer Stem Cells and Provides a Target for Combined Drug Therapy. Cell Stem Cell (2011) 9(5):433–46. doi: 10.1016/j.stem.2011.10.001
175. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The Epithelial-Mesenchymal Transition Generates Cells With Properties of Stem Cells. Cell (2008) 133(4):704–15. doi: 10.1016/j.cell.2008.03.027
176. Lu Y, Lu J, Li X, Zhu H, Fan X, Zhu S, et al. MiR-200a Inhibits Epithelial-Mesenchymal Transition of Pancreatic Cancer Stem Cell. BMC Cancer (2014) 14:85. doi: 10.1186/1471-2407-14-85
177. Thompson EW, Newgreen DF, Tarin D. Carcinoma Invasion and Metastasis: A Role for Epithelial-Mesenchymal Transition? Cancer Res (2005) 65(14):5991–5. doi: 10.1158/0008-5472.CAN-05-0616
178. Thiery JP, Sleeman JP. Complex Networks Orchestrate Epithelial-Mesenchymal Transitions. Nat Rev Mol Cell Biol (2006) 7(2):131–42. doi: 10.1038/nrm1835
179. Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct Regulation of TWIST by HIF-1alpha Promotes Metastasis. Nat Cell Biol (2008) 10(3):295–305. doi: 10.1038/ncb1691
180. Gort EH, van Haaften G, Verlaan I, Groot AJ, Plasterk RH, Shvarts A, et al. The TWIST1 Oncogene is a Direct Target of Hypoxia-Inducible Factor-2alpha. Oncogene (2008) 27(11):1501–10. doi: 10.1038/sj.onc.1210795
181. Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VW, et al. VHL Promotes E2 Box-Dependent E-cadherin Transcription by HIF-mediated Regulation of SIP1 and Snail. Mol Cell Biol (2007) 27(1):157–69. doi: 10.1128/MCB.00892-06
182. Joseph JV, Conroy S, Tomar T, Eggens-Meijer E, Bhat K, Copray S, et al. TGF-Beta is an Inducer of ZEB1-dependent Mesenchymal Transdifferentiation in Glioblastoma That is Associated With Tumor Invasion. Cell Death Dis (2014) 5:e1443. doi: 10.1038/cddis.2014.395
183. Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, et al. EMT and Dissemination Precede Pancreatic Tumor Formation. Cell (2012) 148(1-2):349–61. doi: 10.1016/j.cell.2011.11.025
184. Li Y, Kong D, Ahmad A, Bao B, Sarkar FH. Pancreatic Cancer Stem Cells: Emerging Target for Designing Novel Therapy. Cancer Lett (2013) 338(1):94–100. doi: 10.1016/j.canlet.2012.03.018
185. Cheung KS, Chen L, Chan EW, Seto WK, Wong ICK, Leung WK. Nonsteroidal Anti-Inflammatory Drugs But Not Aspirin are Associated With a Lower Risk of Post-Colonoscopy Colorectal Cancer. Aliment Pharmacol Ther (2020) 51(9):899–908. doi: 10.1111/apt.15693
186. DuBois RN, Giardiello FM, Smalley WE. Nonsteroidal Anti-Inflammatory Drugs, Eicosanoids, and Colorectal Cancer Prevention. Gastroenterol Clin North Am (1996) 25(4):773–91. doi: 10.1016/s0889-8553(05)70274-0
187. Liu Y, Borchert GL, Surazynski A, Phang JM. Proline Oxidase, a p53-induced Gene, Targets COX-2/PGE2 Signaling to Induce Apoptosis and Inhibit Tumor Growth in Colorectal Cancers. Oncogene (2008) 27(53):6729–37. doi: 10.1038/onc.2008.322
188. Zhu Y, Shi C, Zeng L, Liu G, Jiang W, Zhang X, et al. High COX-2 Expression in Cancer-Associated Fibiroblasts Contributes to Poor Survival and Promotes Migration and Invasiveness in Nasopharyngeal Carcinoma. Mol Carcinog (2020) 59(3):265–80. doi: 10.1002/mc.23150
189. Kabir TD, Leigh RJ, Tasena H, Mellone M, Coletta RD, Parkinson EK, et al. A miR-335/COX-2/PTEN Axis Regulates the Secretory Phenotype of Senescent Cancer-Associated Fibroblasts. Aging (Albany NY) (2016) 8(8):1608–35. doi: 10.18632/aging.100987
190. Li T, Yang Y, Hua X, Wang G, Liu W, Jia C, et al. Hepatocellular Carcinoma-Associated Fibroblasts Trigger NK Cell Dysfunction Via PGE2 and IDO. Cancer Lett (2012) 318(2):154–61. doi: 10.1016/j.canlet.2011.12.020
191. Erdogan B, Webb DJ. Cancer-Associated Fibroblasts Modulate Growth Factor Signaling and Extracellular Matrix Remodeling to Regulate Tumor Metastasis. Biochem Soc Trans (2017) 45(1):229–36. doi: 10.1042/BST20160387
192. Herrera M, Herrera A, Dominguez G, Silva J, Garcia V, Garcia JM, et al. Cancer-Associated Fibroblast and M2 Macrophage Markers Together Predict Outcome in Colorectal Cancer Patients. Cancer Sci (2013) 104(4):437–44. doi: 10.1111/cas.12096
193. Katoh H, Watanabe M. Myeloid-Derived Suppressor Cells and Therapeutic Strategies in Cancer. Mediators Inflamm (2015) 2015:159269. doi: 10.1155/2015/159269
194. Pinchuk IV, Beswick EJ, Saada JI, Boya G, Schmitt D, Raju GS, et al. Human Colonic Myofibroblasts Promote Expansion of CD4+ Cd25high Foxp3+ Regulatory T Cells. Gastroenterology (2011) 140(7):2019–30. doi: 10.1053/j.gastro.2011.02.059
195. Inoue C, Miki Y, Saito R, Hata S, Abe J, Sato I, et al. Pd-L1 Induction by Cancer-Associated Fibroblast-Derived Factors in Lung Adenocarcinoma Cells. Cancers (Basel) (2019) 11(9):1257. doi: 10.3390/cancers11091257
196. Ziani L, Chouaib S, Thiery J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front Immunol (2018) 9:414. doi: 10.3389/fimmu.2018.00414
197. Donnarumma E, Fiore D, Nappa M, Roscigno G, Adamo A, Iaboni M, et al. Cancer-Associated Fibroblasts Release Exosomal microRNAs That Dictate an Aggressive Phenotype in Breast Cancer. Oncotarget (2017) 8(12):19592–608. doi: 10.18632/oncotarget.14752
198. Bhome R, Goh RW, Bullock MD, Pillar N, Thirdborough SM, Mellone M, et al. Exosomal microRNAs Derived From Colorectal Cancer-Associated Fibroblasts: Role in Driving Cancer Progression. Aging (Albany NY) (2017) 9(12):2666–94. doi: 10.18632/aging.101355
199. Hu Y, Yan C, Mu L, Huang K, Li X, Tao D, et al. Fibroblast-Derived Exosomes Contribute to Chemoresistance Through Priming Cancer Stem Cells in Colorectal Cancer. PloS One (2015) 10(5):e0125625. doi: 10.1371/journal.pone.0125625
200. Lotti F, Jarrar AM, Pai RK, Hitomi M, Lathia J, Mace A, et al. Chemotherapy Activates Cancer-Associated Fibroblasts to Maintain Colorectal Cancer-Initiating Cells by IL-17A. J Exp Med (2013) 210(13):2851–72. doi: 10.1084/jem.20131195
201. Neri E, Faggioni L, Cerri F, Turini F, Angeli S, Cini L, et al. CT Colonography Versus Double-Contrast Barium Enema for Screening of Colorectal Cancer: Comparison of Radiation Burden. Abdom Imaging (2010) 35(5):596–601. doi: 10.1007/s00261-009-9568-x
202. Brenner H, Chang-Claude J, Jansen L, Knebel P, Stock C, Hoffmeister M. Reduced Risk of Colorectal Cancer Up to 10 Years After Screening, Surveillance, or Diagnostic Colonoscopy. Gastroenterology (2014) 146(3):709–17. doi: 10.1053/j.gastro.2013.09.001
203. Ozguroglu M, Turna H. Do We Really Benefit From Checking Tumor Markers in Detecting Recurrence in Gastrointestinal Cancer? J BUON (2004) 9(2):179–82.
204. Duffy MJ. Tumor Markers in Clinical Practice: A Review Focusing on Common Solid Cancers. Med Princ Pract (2013) 22(1):4–11. doi: 10.1159/000338393
205. van Veelen CW, Verbiest H, Zulch KJ, van Ketel B, van der Vlist MJ, Vlug AM, et al. [Pyruvate Kinase as a Marker of Human Brain Tumors]. Ned Tijdschr Geneeskd (1980) 124(40):1678–85.
206. Mazurek S, Boschek CB, Hugo F, Eigenbrodt E. Pyruvate Kinase Type M2 and its Role in Tumor Growth and Spreading. Semin Cancer Biol (2005) 15(4):300–8. doi: 10.1016/j.semcancer.2005.04.009
207. Eigenbrodt E, Reinacher M, Scheefers-Borchel U, Scheefers H, Friis R. Double Role for Pyruvate Kinase Type M2 in the Expansion of Phosphometabolite Pools Found in Tumor Cells. Crit Rev Oncog (1992) 3(1-2):91–115.
208. Kobierzycki C, Piotrowska A, Latkowski K, Zabel M, Nowak-Markwitz E, Spaczynski M, et al. Correlation of Pyruvate Kinase M2 Expression With Clinicopathological Data in Ovarian Cancer. Anticancer Res (2018) 38(1):295–300. doi: 10.21873/anticanres.12221
209. Liu B, Song M, Qin H, Zhang B, Liu Y, Sun Y, et al. Phosphoribosyl Pyrophosphate Amidotransferase Promotes the Progression of Thyroid Cancer Via Regulating Pyruvate Kinase M2. Onco Targets Ther (2020) 13:7629–39. doi: 10.2147/OTT.S253137
210. Yang Y, Wu K, Liu Y, Shi L, Tao K, Wang G. Prognostic Significance of Metabolic Enzyme Pyruvate Kinase M2 in Breast Cancer: A Meta-Analysis. Med (Baltimore) (2017) 96(46):e8690. doi: 10.1097/MD.0000000000008690
211. Zhu Q, Hong B, Zhang L, Wang J. Pyruvate Kinase M2 Inhibits the Progression of Bladder Cancer by Targeting MAKP Pathway. J Cancer Res Ther (2018) 14(Supplement):S616–S21. doi: 10.4103/0973-1482.187302
212. Guo W, Zhang Z, Li G, Lai X, Gu R, Xu W, et al. Pyruvate Kinase M2 Promotes Prostate Cancer Metastasis Through Regulating ERK1/2-COX-2 Signaling. Front Oncol (2020) 10:544288. doi: 10.3389/fonc.2020.544288
213. Wang C, Zhang S, Liu J, Tian Y, Ma B, Xu S, et al. Secreted Pyruvate Kinase M2 Promotes Lung Cancer Metastasis Through Activating the Integrin Beta1/Fak Signaling Pathway. Cell Rep (2020) 30(6):1780–97.e6. doi: 10.1016/j.celrep.2020.01.037
214. Landt S, Jeschke S, Koeninger A, Thomas A, Heusner T, Korlach S, et al. Tumor-Specific Correlation of Tumor M2 Pyruvate Kinase in Pre-Invasive, Invasive and Recurrent Cervical Cancer. Anticancer Res (2010) 30(2):375–81.
215. Wechsel HW, Petri E, Bichler KH, Feil G. Marker for Renal Cell Carcinoma (RCC): The Dimeric Form of Pyruvate Kinase Type M2 (Tu M2-Pk). Anticancer Res (1999) 19(4A):2583–90.
216. Kumar Y, Tapuria N, Kirmani N, Davidson BR. Tumour M2-pyruvate Kinase: A Gastrointestinal Cancer Marker. Eur J Gastroenterol Hepatol (2007) 19(3):265–76. doi: 10.1097/MEG.0b013e3280102f78
217. Hardt PD, Ewald N. Tumor M2 Pyruvate Kinase: A Tumor Marker and its Clinical Application in Gastrointestinal Malignancy. Expert Rev Mol Diagn (2008) 8(5):579–85. doi: 10.1586/14737159.8.5.579
218. Pantel K, Speicher MR. The Biology of Circulating Tumor Cells. Oncogene (2016) 35(10):1216–24. doi: 10.1038/onc.2015.192
219. Toyoshima K, Hayashi A, Kashiwagi M, Hayashi N, Iwatsuki M, Ishimoto T, et al. Analysis of Circulating Tumor Cells Derived From Advanced Gastric Cancer. Int J Cancer (2015) 137(4):991–8. doi: 10.1002/ijc.29455
220. Witzig TE, Bossy B, Kimlinger T, Roche PC, Ingle JN, Grant C, et al. Detection of Circulating Cytokeratin-Positive Cells in the Blood of Breast Cancer Patients Using Immunomagnetic Enrichment and Digital Microscopy. Clin Cancer Res (2002) 8(5):1085–91.
221. Baccelli I, Schneeweiss A, Riethdorf S, Stenzinger A, Schillert A, Vogel V, et al. Identification of a Population of Blood Circulating Tumor Cells From Breast Cancer Patients That Initiates Metastasis in a Xenograft Assay. Nat Biotechnol (2013) 31(6):539–44. doi: 10.1038/nbt.2576
222. Uenosono Y, Arigami T, Kozono T, Yanagita S, Hagihara T, Haraguchi N, et al. Clinical Significance of Circulating Tumor Cells in Peripheral Blood From Patients With Gastric Cancer. Cancer (2013) 119(22):3984–91. doi: 10.1002/cncr.28309
223. Tang L, Zhao S, Liu W, Parchim NF, Huang J, Tang Y, et al. Diagnostic Accuracy of Circulating Tumor Cells Detection in Gastric Cancer: Systematic Review and Meta-Analysis. BMC Cancer (2013) 13:314. doi: 10.1186/1471-2407-13-314
224. Chen CJ, Sung WW, Chen HC, Chern YJ, Hsu HT, Lin YM, et al. Early Assessment of Colorectal Cancer by Quantifying Circulating Tumor Cells in Peripheral Blood: ECT2 in Diagnosis of Colorectal Cancer. Int J Mol Sci (2017) 18(4):743. doi: 10.3390/ijms18040743
225. Yang C, Shi D, Wang S, Wei C, Zhang C, Xiong B. Prognostic Value of Pre- and Post-Operative Circulating Tumor Cells Detection in Colorectal Cancer Patients Treated With Curative Resection: A Prospective Cohort Study Based on ISET Device. Cancer Manag Res (2018) 10:4135–44. doi: 10.2147/CMAR.S176575
226. Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Koch M, et al. Identification of Double-Stranded Genomic DNA Spanning All Chromosomes With Mutated KRAS and P53 DNA in the Serum Exosomes of Patients With Pancreatic Cancer. J Biol Chem (2014) 289(7):3869–75. doi: 10.1074/jbc.C113.532267
227. Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid Biopsies Come of Age: Towards Implementation of Circulating Tumour DNA. Nat Rev Cancer (2017) 17(4):223–38. doi: 10.1038/nrc.2017.7
228. Sumbal S, Javed A, Afroze B, Zulfiqar HF, Javed F, Noreen S, et al. Circulating Tumor DNA in Blood: Future Genomic Biomarkers for Cancer Detection. Exp Hematol (2018) 65:17–28. doi: 10.1016/j.exphem.2018.06.003
229. Newman AM, Lovejoy AF, Klass DM, Kurtz DM, Chabon JJ, Scherer F, et al. Integrated Digital Error Suppression for Improved Detection of Circulating Tumor DNA. Nat Biotechnol (2016) 34(5):547–55. doi: 10.1038/nbt.3520
230. Leary RJ, Sausen M, Kinde I, Papadopoulos N, Carpten JD, Craig D, et al. Detection of Chromosomal Alterations in the Circulation of Cancer Patients With Whole-Genome Sequencing. Sci Transl Med (2012) 4(162):162ra54. doi: 10.1126/scitranslmed.3004742
231. Haber DA, Velculescu VE. Blood-Based Analyses of Cancer: Circulating Tumor Cells and Circulating Tumor DNA. Cancer Discov (2014) 4(6):650–61. doi: 10.1158/2159-8290.CD-13-1014
232. Kidess E, Jeffrey SS. Circulating Tumor Cells Versus Tumor-Derived Cell-Free DNA: Rivals or Partners in Cancer Care in the Era of Single-Cell Analysis? Genome Med (2013) 5(8):70. doi: 10.1186/gm474
233. Fiala C, Diamandis EP. Utility of Circulating Tumor DNA in Cancer Diagnostics With Emphasis on Early Detection. BMC Med (2018) 16(1):166. doi: 10.1186/s12916-018-1157-9
234. Wang HQ, Yang CY, Wang SY, Wang T, Han JL, Wei K, et al. Cell-Free Plasma Hypermethylated CASZ1, CDH13 and ING2 are Promising Biomarkers of Esophageal Cancer. J BioMed Res (2018) 32(5):424–33. doi: 10.7555/JBR.32.20170065
235. Tian X, Sun B, Chen C, Gao C, Zhang J, Lu X, et al. Circulating Tumor DNA 5-Hydroxymethylcytosine as a Novel Diagnostic Biomarker for Esophageal Cancer. Cell Res (2018) 28(5):597–600. doi: 10.1038/s41422-018-0014-x
236. Qian C, Ju S, Qi J, Zhao J, Shen X, Jing R, et al. Alu-Based Cell-Free DNA: A Novel Biomarker for Screening of Gastric Cancer. Oncotarget (2017) 8(33):54037–45. doi: 10.18632/oncotarget.11079
237. Gao Y, Zhang K, Xi H, Cai A, Wu X, Cui J, et al. Diagnostic and Prognostic Value of Circulating Tumor DNA in Gastric Cancer: A Meta-Analysis. Oncotarget (2017) 8(4):6330–40. doi: 10.18632/oncotarget.14064
238. Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, et al. Circulating Tumour DNA Methylation Markers for Diagnosis and Prognosis of Hepatocellular Carcinoma. Nat Mater (2017) 16(11):1155–61. doi: 10.1038/nmat4997
239. Tao K, Bian Z, Zhang Q, Guo X, Yin C, Wang Y, et al. Machine Learning-Based Genome-Wide Interrogation of Somatic Copy Number Aberrations in Circulating Tumor DNA for Early Detection of Hepatocellular Carcinoma. EBioMedicine (2020) 56:102811. doi: 10.1016/j.ebiom.2020.102811
240. Jaworski JJ, Morgan RD, Sivakumar S. Circulating Cell-Free Tumour DNA for Early Detection of Pancreatic Cancer. Cancers (Basel) (2020) 12(12):3704. doi: 10.3390/cancers12123704
241. Grunvald MW, Jacobson RA, Kuzel TM, Pappas SG, Masood A. Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer. Int J Mol Sci (2020) 21(20):7651. doi: 10.3390/ijms21207651
242. Liggett T, Melnikov A, Yi QL, Replogle C, Brand R, Kaul K, et al. Differential Methylation of Cell-Free Circulating DNA Among Patients With Pancreatic Cancer Versus Chronic Pancreatitis. Cancer (2010) 116(7):1674–80. doi: 10.1002/cncr.24893
243. Eissa MAL, Lerner L, Abdelfatah E, Shankar N, Canner JK, Hasan NM, et al. Promoter Methylation of ADAMTS1 and BNC1 as Potential Biomarkers for Early Detection of Pancreatic Cancer in Blood. Clin Epigenet (2019) 11(1):59. doi: 10.1186/s13148-019-0650-0
244. Cohen JD, Javed AA, Thoburn C, Wong F, Tie J, Gibbs P, et al. Combined Circulating Tumor DNA and Protein Biomarker-Based Liquid Biopsy for the Earlier Detection of Pancreatic Cancers. Proc Natl Acad Sci USA (2017) 114(38):10202–7. doi: 10.1073/pnas.1704961114
245. Bedin C, Enzo MV, Del Bianco P, Pucciarelli S, Nitti D, Agostini M. Diagnostic and Prognostic Role of Cell-Free DNA Testing for Colorectal Cancer Patients. Int J Cancer (2017) 140(8):1888–98. doi: 10.1002/ijc.30565
246. Nagai Y, Sunami E, Yamamoto Y, Hata K, Okada S, Murono K, et al. LINE-1 Hypomethylation Status of Circulating Cell-Free DNA in Plasma as a Biomarker for Colorectal Cancer. Oncotarget (2017) 8(7):11906–16. doi: 10.18632/oncotarget.14439
247. Brennecke J, Stark A, Russell RB, Cohen SM. Principles of microRNA-target Recognition. PloS Biol (2005) 3(3):e85. doi: 10.1371/journal.pbio.0030085
248. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. The microRNA Spectrum in 12 Body Fluids. Clin Chem (2010) 56(11):1733–41. doi: 10.1373/clinchem.2010.147405
249. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent Deletions and Down-Regulation of Micro- RNA Genes miR15 and miR16 at 13q14 in Chronic Lymphocytic Leukemia. Proc Natl Acad Sci USA (2002) 99(24):15524–9. doi: 10.1073/pnas.242606799
250. Shen Y, Ding Y, Ma Q, Zhao L, Guo X, Shao Y, et al. Identification of Novel Circulating Mirna Biomarkers for the Diagnosis of Esophageal Squamous Cell Carcinoma and Squamous Dysplasia. Cancer Epidemiol Biomarkers Prev (2019) 28(7):1212–20. doi: 10.1158/1055-9965.EPI-18-1199
251. Yang M, Liu R, Sheng J, Liao J, Wang Y, Pan E, et al. Differential Expression Profiles of microRNAs as Potential Biomarkers for the Early Diagnosis of Esophageal Squamous Cell Carcinoma. Oncol Rep (2013) 29(1):169–76. doi: 10.3892/or.2012.2105
252. So JBY, Kapoor R, Zhu F, Koh C, Zhou L, Zou R, et al. Development and Validation of a Serum microRNA Biomarker Panel for Detecting Gastric Cancer in a High-Risk Population. Gut (2020) 70(5):829–37. doi: 10.1136/gutjnl-2020-322065
253. Shen S, Lin Y, Yuan X, Shen L, Chen J, Chen L, et al. Biomarker MicroRNAs for Diagnosis, Prognosis and Treatment of Hepatocellular Carcinoma: A Functional Survey and Comparison. Sci Rep (2016) 6:38311. doi: 10.1038/srep38311
254. Shams R, Saberi S, Zali M, Sadeghi A, Ghafouri-Fard S, Aghdaei HA. Identification of Potential microRNA Panels for Pancreatic Cancer Diagnosis Using Microarray Datasets and Bioinformatics Methods. Sci Rep (2020) 10(1):7559. doi: 10.1038/s41598-020-64569-1
255. Eslamizadeh S, Heidari M, Agah S, Faghihloo E, Ghazi H, Mirzaei A, et al. The Role of MicroRNA Signature as Diagnostic Biomarkers in Different Clinical Stages of Colorectal Cancer. Cell J (2018) 20(2):220–30. doi: 10.22074/cellj.2018.5366
256. Li G, Shen Q, Li C, Li D, Chen J, He M. Identification of Circulating MicroRNAs as Novel Potential Biomarkers for Hepatocellular Carcinoma Detection: A Systematic Review and Meta-Analysis. Clin Transl Oncol (2015) 17(9):684–93. doi: 10.1007/s12094-015-1294-y
257. Pan C, Yan X, Li H, Huang L, Yin M, Yang Y, et al. Systematic Literature Review and Clinical Validation of Circulating microRNAs as Diagnostic Biomarkers for Colorectal Cancer. Oncotarget (2017) 8(40):68317–28. doi: 10.18632/oncotarget.19344
258. Nedaeinia R, Manian M, Jazayeri MH, Ranjbar M, Salehi R, Sharifi M, et al. Circulating Exosomes and Exosomal microRNAs as Biomarkers in Gastrointestinal Cancer. Cancer Gene Ther (2017) 24(2):48–56. doi: 10.1038/cgt.2016.77
259. Wang R, Wen H, Xu Y, Chen Q, Luo Y, Lin Y, et al. Circulating microRNAs as a Novel Class of Diagnostic Biomarkers in Gastrointestinal Tumors Detection: A Meta-Analysis Based on 42 Articles. PloS One (2014) 9(11):e113401. doi: 10.1371/journal.pone.0113401
260. Salzman J. Circular RNA Expression: Its Potential Regulation and Function. Trends Genet (2016) 32(5):309–16. doi: 10.1016/j.tig.2016.03.002
261. Wang S, Zhang K, Tan S, Xin J, Yuan Q, Xu H, et al. Circular RNAs in Body Fluids as Cancer Biomarkers: The New Frontier of Liquid Biopsies. Mol Cancer (2021) 20(1):13. doi: 10.1186/s12943-020-01298-z
262. Liu X, Abraham JM, Cheng Y, Wang Z, Wang Z, Zhang G, et al. Synthetic Circular Rna Functions as a Mir-21 Sponge to Suppress Gastric Carcinoma Cell Proliferation. Mol Ther Nucleic Acids (2018) 13:312–21. doi: 10.1016/j.omtn.2018.09.010
263. Niu C, Zhao L, Guo X, Shen Y, Shao Y, Liu F. Diagnostic Accuracy of circRNAs in Esophageal Cancer: A Meta-Analysis. Dis Markers (2019) 2019:9673129. doi: 10.1155/2019/9673129
264. Li T, Shao Y, Fu L, Xie Y, Zhu L, Sun W, et al. Plasma Circular RNA Profiling of Patients With Gastric Cancer and Their Droplet Digital RT-PCR Detection. J Mol Med (Berl) (2018) 96(1):85–96. doi: 10.1007/s00109-017-1600-y
265. Zhao Q, Chen S, Li T, Xiao B, Zhang X. Clinical Values of Circular RNA 0000181 in the Screening of Gastric Cancer. J Clin Lab Anal (2018) 32(4):e22333. doi: 10.1002/jcla.22333
266. Sun XH, Wang YT, Li GF, Zhang N, Fan L. Serum-Derived three-circRNA Signature as a Diagnostic Biomarker for Hepatocellular Carcinoma. Cancer Cell Int (2020) 20:226. doi: 10.1186/s12935-020-01302-y
267. Yang F, Liu DY, Guo JT, Ge N, Zhu P, Liu X, et al. Circular RNA circ-LDLRAD3 as a Biomarker in Diagnosis of Pancreatic Cancer. World J Gastroenterol (2017) 23(47):8345–54. doi: 10.3748/wjg.v23.i47.8345
268. Yao J, Zhang C, Chen Y, Gao S. Downregulation of Circular RNA circ-LDLRAD3 Suppresses Pancreatic Cancer Progression Through miR-137-3p/PTN Axis. Life Sci (2019) 239:116871. doi: 10.1016/j.lfs.2019.116871
269. Lin J, Cai D, Li W, Yu T, Mao H, Jiang S, et al. Plasma Circular RNA Panel Acts as a Novel Diagnostic Biomarker for Colorectal Cancer. Clin Biochem (2019) 74:60–8. doi: 10.1016/j.clinbiochem.2019.10.012
270. Li J, Song Y, Wang J, Huang J. Plasma Circular RNA Panel Acts as a Novel Diagnostic Biomarker for Colorectal Cancer Detection. Am J Transl Res (2020) 12(11):7395–403.
271. Tao Z, Li S, Ichim TE, Yang J, Riordan N, Yenugonda V, et al. Cellular Immunotherapy of Cancer: An Overview and Future Directions. Immunotherapy (2017) 9(7):589–606. doi: 10.2217/imt-2016-0086
272. Rodriguez Perez A, Campillo-Davo D, Van Tendeloo VFI, Benitez-Ribas D. Cellular Immunotherapy: A Clinical State-of-the-Art of a New Paradigm for Cancer Treatment. Clin Transl Oncol (2020) 22(11):1923–37. doi: 10.1007/s12094-020-02344-4
273. Krzyszczyk P, Acevedo A, Davidoff EJ, Timmins LM, Marrero-Berrios I, Patel M, et al. The Growing Role of Precision and Personalized Medicine for Cancer Treatment. Technol (Singap World Sci) (2018) 6(3-4):79–100. doi: 10.1142/S2339547818300020
274. Dumbrava EI, Meric-Bernstam F. Personalized Cancer Therapy-Leveraging a Knowledge Base for Clinical Decision-Making. Cold Spring Harb Mol Case Stud (2018) 4(2):a001578. doi: 10.1101/mcs.a001578
275. McCarthy EF. The Toxins of William B. Coley and the Treatment of Bone and Soft-Tissue Sarcomas. Iowa Orthop J (2006) 26:154–8. doi: 10.1177/104990919100800102
276. Rasmussen L, Arvin A. Chemotherapy-Induced Immunosuppression. Environ Health Perspect (1982) 43:21–5. doi: 10.1289/ehp.824321
277. Steele TA. Chemotherapy-Induced Immunosuppression and Reconstitution of Immune Function. Leuk Res (2002) 26(4):411–4. doi: 10.1016/s0145-2126(01)00138-2
278. Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T, et al. Chemotherapy Induces Programmed Cell Death-Ligand 1 Overexpression Via the Nuclear Factor-Kappab to Foster an Immunosuppressive Tumor Microenvironment in Ovarian Cancer. Cancer Res (2015) 75(23):5034–45. doi: 10.1158/0008-5472.CAN-14-3098
279. Daenen LG, Roodhart JM, van Amersfoort M, Dehnad M, Roessingh W, Ulfman LH, et al. Chemotherapy Enhances Metastasis Formation Via VEGFR-1-expressing Endothelial Cells. Cancer Res (2011) 71(22):6976–85. doi: 10.1158/0008-5472.CAN-11-0627
280. Palmieri LJ, Dubreuil O, Bachet JB, Trouilloud I, Locher C, Coriat R, et al. Reasons for Chemotherapy Discontinuation and End-of-Life in Patients With Gastrointestinal Cancer: A Multicenter Prospective AGEO Study. Clin Res Hepatol Gastroenterol (2021) 45(1):101431. doi: 10.1016/j.clinre.2020.03.029
281. Park SE, Lee SH, Ahn JS, Ahn MJ, Park K, Sun JM. Increased Response Rates to Salvage Chemotherapy Administered After PD-1/PD-L1 Inhibitors in Patients With Non-Small Cell Lung Cancer. J Thorac Oncol (2018) 13(1):106–11. doi: 10.1016/j.jtho.2017.10.011
282. Jiang J, Xu N, Wu C, Deng H, Lu M, Li M, et al. Treatment of Advanced Gastric Cancer by Chemotherapy Combined With Autologous Cytokine-Induced Killer Cells. Anticancer Res (2006) 26(3B):2237–42.
283. Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin Selectively Targets Cancer Stem Cells, and Acts Together With Chemotherapy to Block Tumor Growth and Prolong Remission. Cancer Res (2009) 69(19):7507–11. doi: 10.1158/0008-5472.CAN-09-2994
284. Helleday T. Making Immunotherapy ‘Cold’ Tumours ‘Hot’ by Chemotherapy-Induced Mutations-a Misconception. Ann Oncol (2019) 30(3):360–1. doi: 10.1093/annonc/mdz013
285. Spiegel S, Milstien S, Grant S. Endogenous Modulators and Pharmacological Inhibitors of Histone Deacetylases in Cancer Therapy. Oncogene (2012) 31(5):537–51. doi: 10.1038/onc.2011.267
286. Li G, Tian Y, Zhu WG. The Roles of Histone Deacetylases and Their Inhibitors in Cancer Therapy. Front Cell Dev Biol (2020) 8:576946. doi: 10.3389/fcell.2020.576946
287. Masunaga SI, Sanada Y, Tano K, Sakurai Y, Tanaka H, Takata T, et al. An Attempt to Improve the Therapeutic Effect of Boron Neutron Capture Therapy Using Commonly Employed 10B-Carriers Based on Analytical Studies on the Correlation Among Quiescent Tumor Cell Characteristics, Tumor Heterogeneity and Cancer Stemness. J Radiat Res (2020) 61(6):876–85. doi: 10.1093/jrr/rraa048
288. Hatanaka H, Amano K, Kamano S, Fankhauser H, Hanamura T, Sano K. Boron-Neutron Capture Therapy in Relation to Immunotherapy. Acta Neurochir (Wien) (1978) 42(1-2):57–72. doi: 10.1007/BF01406631
289. Rosenberg SA, Spiess P, Lafreniere R. A New Approach to the Adoptive Immunotherapy of Cancer With Tumor-Infiltrating Lymphocytes. Science (1986) 233(4770):1318–21. doi: 10.1126/science.3489291
290. Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-Infiltrating Lymphocytes: Warriors Fight Against Tumors Powerfully. BioMed Pharmacother (2020) 132:110873. doi: 10.1016/j.biopha.2020.110873
291. D’Andrea MA, Reddy GK. Systemic Effects of Radiation Therapy-Induced Abscopal Responses in Patients With Advanced Lung Cancer. Oncology (2021) 99(1):1–14. doi: 10.1159/000510287
292. Yang C, Li Y, Yang Y, Chen Z. Overview of Strategies to Improve Therapy Against Tumors Using Natural Killer Cell. J Immunol Res (2020) 2020:8459496. doi: 10.1155/2020/8459496
293. Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J. Car-NK Cells: A Promising Cellular Immunotherapy for Cancer. EBioMedicine (2020) 59:102975. doi: 10.1016/j.ebiom.2020.102975
294. Marangon M, Visco C, Barbui AM, Chiappella A, Fabbri A, Ferrero S, et al. Allogeneic Stem Cell Transplantation in Mantle Cell Lymphoma in the Era of New Drugs and CAR-T Cell Therapy. Cancers (Basel) (2021) 13(2):291. doi: 10.3390/cancers13020291
295. Zhao YJ, Jiang N, Song QK, Wu JP, Song YG, Zhang HM, et al. Continuous DC-CIK Infusions Restore CD8+ Cellular Immunity, Physical Activity and Improve Clinical Efficacy in Advanced Cancer Patients Unresponsive to Conventional Treatments. Asian Pac J Cancer Prev (2015) 16(6):2419–23. doi: 10.7314/apjcp.2015.16.6.2419
296. Zhang L, Mu Y, Zhang A, Xie J, Chen S, Xu F, et al. Cytokine-Induced Killer Cells/Dendritic Cells-Cytokine Induced Killer Cells Immunotherapy Combined With Chemotherapy for Treatment of Colorectal Cancer in China: A Meta-Analysis of 29 Trials Involving 2,610 Patients. Oncotarget (2017) 8(28):45164–77. doi: 10.18632/oncotarget.16665
297. Mu Y, Zhou CH, Chen SF, Ding J, Zhang YX, Yang YP, et al. Effectiveness and Safety of Chemotherapy Combined With Cytokine-Induced Killer Cell /Dendritic Cell-Cytokine-Induced Killer Cell Therapy for Treatment of Gastric Cancer in China: A Systematic Review and Meta-Analysis. Cytotherapy (2016) 18(9):1162–77. doi: 10.1016/j.jcyt.2016.05.015
298. Jiang N, Qiao G, Wang X, Morse MA, Gwin WR, Zhou L, et al. Dendritic Cell/Cytokine-Induced Killer Cell Immunotherapy Combined With S-1 in Patients With Advanced Pancreatic Cancer: A Prospective Study. Clin Cancer Res (2017) 23(17):5066–73. doi: 10.1158/1078-0432.CCR-17-0492
299. Cao J, Kong FH, Liu X, Wang XB. Immunotherapy With Dendritic Cells and Cytokine-Induced Killer Cells for Hepatocellular Carcinoma: A Meta-Analysis. World J Gastroenterol (2019) 25(27):3649–63. doi: 10.3748/wjg.v25.i27.3649
300. Merker M, Salzmann-Manrique E, Katzki V, Huenecke S, Bremm M, Bakhtiar S, et al. Clearance of Hematologic Malignancies by Allogeneic Cytokine-Induced Killer Cell or Donor Lymphocyte Infusions. Biol Blood Marrow Transplant (2019) 25(7):1281–92. doi: 10.1016/j.bbmt.2019.03.004
301. Liang S, Xu K, Niu L, Wang X, Liang Y, Zhang M, et al. Comparison of Autogeneic and Allogeneic Natural Killer Cells Immunotherapy on the Clinical Outcome of Recurrent Breast Cancer. Onco Targets Ther (2017) 10:4273–81. doi: 10.2147/OTT.S139986
302. Wang F, Lau JKC, Yu J. The Role of Natural Killer Cell in Gastrointestinal Cancer: Killer or Helper. Oncogene (2020) 40(4):717–30. doi: 10.1038/s41388-020-01561-z
303. Lin M, Liang S, Wang X, Liang Y, Zhang M, Chen J, et al. Percutaneous Irreversible Electroporation Combined With Allogeneic Natural Killer Cell Immunotherapy for Patients With Unresectable (Stage III/IV) Pancreatic Cancer: A Promising Treatment. J Cancer Res Clin Oncol (2017) 143(12):2607–18. doi: 10.1007/s00432-017-2513-4
304. Yang Y, Qin Z, Du D, Wu Y, Qiu S, Mu F, et al. Safety and Short-Term Efficacy of Irreversible Electroporation and Allogenic Natural Killer Cell Immunotherapy Combination in the Treatment of Patients With Unresectable Primary Liver Cancer. Cardiovasc Intervent Radiol (2019) 42(1):48–59. doi: 10.1007/s00270-018-2069-y
305. Lim TS, Chew V, Sieow JL, Goh S, Yeong JP, Soon AL, et al. PD-1 Expression on Dendritic Cells Suppresses CD8(+) T Cell Function and Antitumor Immunity. Oncoimmunology (2016) 5(3):e1085146. doi: 10.1080/2162402X.2015.1085146
306. Peng Q, Qiu X, Zhang Z, Zhang S, Zhang Y, Liang Y, et al. Pd-L1 on Dendritic Cells Attenuates T Cell Activation and Regulates Response to Immune Checkpoint Blockade. Nat Commun (2020) 11(1):4835. doi: 10.1038/s41467-020-18570-x
307. Vari F, Arpon D, Keane C, Hertzberg MS, Talaulikar D, Jain S, et al. Immune Evasion Via PD-1/PD-L1 on NK Cells and Monocyte/Macrophages is More Prominent in Hodgkin Lymphoma Than DLBCL. Blood (2018) 131(16):1809–19. doi: 10.1182/blood-2017-07-796342
308. Norris S, Coleman A, Kuri-Cervantes L, Bower M, Nelson M, Goodier MR. PD-1 Expression on Natural Killer Cells and CD8(+) T Cells During Chronic HIV-1 Infection. Viral Immunol (2012) 25(4):329–32. doi: 10.1089/vim.2011.0096
309. Mariotti FR, Petrini S, Ingegnere T, Tumino N, Besi F, Scordamaglia F, et al. PD-1 in Human NK Cells: Evidence of Cytoplasmic mRNA and Protein Expression. Oncoimmunology (2019) 8(3):1557030. doi: 10.1080/2162402X.2018.1557030
310. Beldi-Ferchiou A, Lambert M, Dogniaux S, Vely F, Vivier E, Olive D, et al. PD-1 Mediates Functional Exhaustion of Activated NK Cells in Patients With Kaposi Sarcoma. Oncotarget (2016) 7(45):72961–77. doi: 10.18632/oncotarget.12150
311. Niu C, Li M, Zhu S, Chen Y, Zhou L, Xu D, et al. PD-1-Positive Natural Killer Cells Have a Weaker Antitumor Function Than That of PD-1-negative Natural Killer Cells in Lung Cancer. Int J Med Sci (2020) 17(13):1964–73. doi: 10.7150/ijms.47701
312. Concha-Benavente F, Kansy B, Moskovitz J, Moy J, Chandran U, Ferris RL. Pd-L1 Mediates Dysfunction in Activated Pd-1(+) NK Cells in Head and Neck Cancer Patients. Cancer Immunol Res (2018) 6(12):1548–60. doi: 10.1158/2326-6066.CIR-18-0062
313. Giuliani M, Janji B, Berchem G. Activation of NK Cells and Disruption of PD-L1/PD-1 Axis: Two Different Ways for Lenalidomide to Block Myeloma Progression. Oncotarget (2017) 8(14):24031–44. doi: 10.18632/oncotarget.15234
314. Zhang W, Song Z, Xiao J, Liu X, Luo Y, Yang Z, et al. Blocking the PD-1/PD-L1 Axis in Dendritic Cell-Stimulated Cytokine-Induced Killer Cells With Pembrolizumab Enhances Their Therapeutic Effects Against Hepatocellular Carcinoma. J Cancer (2019) 10(11):2578–87. doi: 10.7150/jca.26961
315. Oyer JL, Gitto SB, Altomare DA, Copik AJ. Pd-L1 Blockade Enhances Anti-Tumor Efficacy of NK Cells. Oncoimmunology (2018) 7(11):e1509819. doi: 10.1080/2162402X.2018.1509819
316. Itzhaki O, Jacoby E, Nissani A, Levi M, Nagler A, Kubi A, et al. Head-to-Head Comparison of in-House Produced CD19 CAR-T Cell in ALL and NHL Patients. J Immunother Cancer (2020) 8(1):e000148. doi: 10.1136/jitc-2019-000148
317. Li J, Li W, Huang K, Zhang Y, Kupfer G, Zhao Q. Chimeric Antigen Receptor T Cell (CAR-T) Immunotherapy for Solid Tumors: Lessons Learned and Strategies for Moving Forward. J Hematol Oncol (2018) 11(1):22. doi: 10.1186/s13045-018-0568-6
318. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DA, Feldman SA, et al. T Cells Targeting Carcinoembryonic Antigen can Mediate Regression of Metastatic Colorectal Cancer But Induce Severe Transient Colitis. Mol Ther (2011) 19(3):620–6. doi: 10.1038/mt.2010.272
319. Sanmamed MF, Chen L. A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell (2019) 176(3):677. doi: 10.1016/j.cell.2019.01.008
320. Tang X, Yang L, Li Z, Nalin AP, Dai H, Xu T, et al. First-in-Man Clinical Trial of CAR Nk-92 Cells: Safety Test of CD33-CAR Nk-92 Cells in Patients With Relapsed and Refractory Acute Myeloid Leukemia. Am J Cancer Res (2018) 8(6):1083–9.
321. Hollingsworth RE, Jansen K. Turning the Corner on Therapeutic Cancer Vaccines. NPJ Vaccines (2019) 4:7. doi: 10.1038/s41541-019-0103-y
322. Leitner WW, Ying H, Restifo NP. DNA and RNA-based Vaccines: Principles, Progress and Prospects. Vaccine (1999) 18(9-10):765–77. doi: 10.1016/s0264-410x(99)00271-6
323. Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, et al. Safety, Efficacy, and Immunogenicity of VGX-3100, a Therapeutic Synthetic DNA Vaccine Targeting Human Papillomavirus 16 and 18 E6 and E7 Proteins for Cervical Intraepithelial Neoplasia 2/3: A Randomised, Double-Blind, Placebo-Controlled Phase 2b Trial. Lancet (2015) 386(10008):2078–88. doi: 10.1016/S0140-6736(15)00239-1
324. Norell H, Poschke I, Charo J, Wei WZ, Erskine C, Piechocki MP, et al. Vaccination With a Plasmid DNA Encoding HER-2/neu Together With Low Doses of GM-CSF and IL-2 in Patients With Metastatic Breast Carcinoma: A Pilot Clinical Trial. J Transl Med (2010) 8:53. doi: 10.1186/1479-5876-8-53
325. Staff C, Mozaffari F, Haller BK, Wahren B, Liljefors M. A Phase I Safety Study of Plasmid DNA Immunization Targeting Carcinoembryonic Antigen in Colorectal Cancer Patients. Vaccine (2011) 29(39):6817–22. doi: 10.1016/j.vaccine.2010.12.063
326. Chudley L, McCann K, Mander A, Tjelle T, Campos-Perez J, Godeseth R, et al. DNA Fusion-Gene Vaccination in Patients With Prostate Cancer Induces High-Frequency CD8(+) T-Cell Responses and Increases PSA Doubling Time. Cancer Immunol Immunother (2012) 61(11):2161–70. doi: 10.1007/s00262-012-1270-0
327. Ginsberg BA, Gallardo HF, Rasalan TS, Adamow M, Mu Z, Tandon S, et al. Immunologic Response to Xenogeneic Gp100 DNA in Melanoma Patients: Comparison of Particle-Mediated Epidermal Delivery With Intramuscular Injection. Clin Cancer Res (2010) 16(15):4057–65. doi: 10.1158/1078-0432.CCR-10-1093
328. Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, et al. mRNA Vaccine-Induced Neoantigen-Specific T Cell Immunity in Patients With Gastrointestinal Cancer. J Clin Invest (2020) 130(11):5976–88. doi: 10.1172/JCI134915
329. Sadanaga N, Nagashima H, Mashino K, Tahara K, Yamaguchi H, Ohta M, et al. Dendritic Cell Vaccination With MAGE Peptide is a Novel Therapeutic Approach for Gastrointestinal Carcinomas. Clin Cancer Res (2001) 7(8):2277–84.
330. Lion E, Smits EL, Berneman ZN, Van Tendeloo VF. NK Cells: Key to Success of DC-based Cancer Vaccines? Oncologist (2012) 17(10):1256–70. doi: 10.1634/theoncologist.2011-0122
331. Hammerich L, Binder A, Brody JD. In Situ Vaccination: Cancer Immunotherapy Both Personalized and Off-the-Shelf. Mol Oncol (2015) 9(10):1966–81. doi: 10.1016/j.molonc.2015.10.016
332. Sheen MR, Fiering S. In Situ Vaccination: Harvesting Low Hanging Fruit on the Cancer Immunotherapy Tree. Wiley Interdiscip Rev Nanomed Nanobiotechnol (2019) 11(1):e1524. doi: 10.1002/wnan.1524
333. Cebon J. Perspective: Cancer Vaccines in the Era of Immune Checkpoint Blockade. Mamm Genome (2018) 29(11-12):703–13. doi: 10.1007/s00335-018-9786-z
334. Mougel A, Terme M, Tanchot C. Therapeutic Cancer Vaccine and Combinations With Antiangiogenic Therapies and Immune Checkpoint Blockade. Front Immunol (2019) 10:467. doi: 10.3389/fimmu.2019.00467
335. Stojdl DF, Lichty B, Knowles S, Marius R, Atkins H, Sonenberg N, et al. Exploiting Tumor-Specific Defects in the Interferon Pathway With a Previously Unknown Oncolytic Virus. Nat Med (2000) 6(7):821–5. doi: 10.1038/77558
336. Coffey MC, Strong JE, Forsyth PA, Lee PW. Reovirus Therapy of Tumors With Activated Ras Pathway. Science (1998) 282(5392):1332–4. doi: 10.1126/science.282.5392.1332
337. Yokoda RT, Nagalo BM, Borad MJ. Oncolytic Adenoviruses in Gastrointestinal Cancers. Biomedicines (2018) 6(1):33. doi: 10.3390/biomedicines6010033
338. Rahman MM, McFadden G. Oncolytic Virotherapy With Myxoma Virus. J Clin Med (2020) 9(1):171. doi: 10.3390/jcm9010171
339. Abate-Daga D, Andreu N, Camacho-Sanchez J, Alemany R, Herance R, Millan O, et al. Oncolytic Adenoviruses Armed With Thymidine Kinase can be Traced by PET Imaging and Show Potent Antitumoural Effects by Ganciclovir Dosing. PloS One (2011) 6(10):e26142. doi: 10.1371/journal.pone.0026142
340. Galanis E, Atherton PJ, Maurer MJ, Knutson KL, Dowdy SC, Cliby WA, et al. Oncolytic Measles Virus Expressing the Sodium Iodide Symporter to Treat Drug-Resistant Ovarian Cancer. Cancer Res (2015) 75(1):22–30. doi: 10.1158/0008-5472.CAN-14-2533
341. McCart JA, Mehta N, Scollard D, Reilly RM, Carrasquillo JA, Tang N, et al. Oncolytic Vaccinia Virus Expressing the Human Somatostatin Receptor SSTR2: Molecular Imaging After Systemic Delivery Using 111In-Pentetreotide. Mol Ther (2004) 10(3):553–61. doi: 10.1016/j.ymthe.2004.06.158
342. Barton KN, Stricker H, Brown SL, Elshaikh M, Aref I, Lu M, et al. Phase I Study of Noninvasive Imaging of Adenovirus-Mediated Gene Expression in the Human Prostate. Mol Ther (2008) 16(10):1761–9. doi: 10.1038/mt.2008.172
343. Jacobs A, Tjuvajev JG, Dubrovin M, Akhurst T, Balatoni J, Beattie B, et al. Positron Emission Tomography-Based Imaging of Transgene Expression Mediated by Replication-Conditional, Oncolytic Herpes Simplex Virus Type 1 Mutant Vectors In Vivo. Cancer Res (2001) 61(7):2983–95.
344. Zellmer VR, Zhang S. Evolving Concepts of Tumor Heterogeneity. Cell Biosci (2014) 4:69. doi: 10.1186/2045-3701-4-69
345. McGranahan N, Swanton C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell (2017) 168(4):613–28. doi: 10.1016/j.cell.2017.01.018
346. Parikh AR, Leshchiner I, Elagina L, Goyal L, Levovitz C, Siravegna G, et al. Liquid Versus Tissue Biopsy for Detecting Acquired Resistance and Tumor Heterogeneity in Gastrointestinal Cancers. Nat Med (2019) 25(9):1415–21. doi: 10.1038/s41591-019-0561-9
347. Aravanis AM, Lee M, Klausner RD. Next-Generation Sequencing of Circulating Tumor DNA for Early Cancer Detection. Cell (2017) 168(4):571–4. doi: 10.1016/j.cell.2017.01.030
348. Hass R, von der Ohe J, Ungefroren H. Impact of the Tumor Microenvironment on Tumor Heterogeneity and Consequences for Cancer Cell Plasticity and Stemness. Cancers (Basel) (2020) 12(12):3716. doi: 10.3390/cancers12123716
349. Yuan Y. Spatial Heterogeneity in the Tumor Microenvironment. Cold Spring Harb Perspect Med (2016) 6(8):a026583. doi: 10.1101/cshperspect.a026583
350. Cell Editorial T. Cancer: The Road Ahead. Cell (2017) 168(4):545–6. doi: 10.1016/j.cell.2017.01.036
351. El-Deiry WS, Taylor B, Neal JW. Tumor Evolution, Heterogeneity, and Therapy for Our Patients With Advanced Cancer: How Far Have We Come? Am Soc Clin Oncol Educ Book (2017) 37:e8–15. doi: 10.14694/EDBK_175524
Keywords: gastrointestinal cancers, tumor microenvironment, tumorigenesis, early detection, cellular immunotherapy
Citation: Zhu H and Liu X (2021) Advances of Tumorigenesis, Diagnosis at Early Stage, and Cellular Immunotherapy in Gastrointestinal Malignancies. Front. Oncol. 11:666340. doi: 10.3389/fonc.2021.666340
Received: 10 February 2021; Accepted: 19 July 2021;
Published: 09 August 2021.
Edited by:
Ruowen Zhang, Stony Brook University, United StatesReviewed by:
Sheng Li, Shandong Tumor Hospital, ChinaShuiling Jin, First Affiliated Hosptital of Zhengzhou University, China
Copyright © 2021 Zhu and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Haipeng Zhu, dr.haipengzhu@cimingboao.com; dr.haipengzhu@hotmail.com