Open access peer-reviewed chapter - ONLINE FIRST

New Approaches in Gastric Cancer Immunotherapy

Written By

Pegah Mousavi, Ali Ahmadi, Shakila Behzadifar, Javad Mohammadnejad and Seyed Mohammad Hosseini

Submitted: 01 March 2024 Reviewed: 11 March 2024 Published: 01 July 2024

DOI: 10.5772/intechopen.1005783

Gastric Cancer - Progress and Challenges in the Era of Precision Medicine IntechOpen
Gastric Cancer - Progress and Challenges in the Era of Precision ... Edited by Daniela Lazar

From the Edited Volume

Gastric Cancer - Progress and Challenges in the Era of Precision Medicine [Working Title]

Dr. Daniela Cornelia Lazar

Chapter metrics overview

3 Chapter Downloads

View Full Metrics

Abstract

Cancer has an inferior prognosis in most cases and is often challenging to treat. Gastric cancer (GC), which is among leading causes of the top five malignant tumor deaths worldwide and whose incidence is increasing every day, is no exception. GC is frequently diagnosed at a progressive or metastatic stage of the disease. At this stage, the clinical effectiveness of conventional treatments such as surgery and chemotherapy is limited, and the median overall survival is reduced to only about a few months. The tumor microenvironment (TME) and the specific conditions that govern it, concurrently with multiple mutations, have significantly increased the resistance of cancer cells. However, the study of molecular biology, cell signaling pathways, and immune system function provides a new approach using immunotherapy such as immune inhibitors, T cell transfer therapy, monoclonal antibodies (mAbs), therapeutic vaccines, etc. to overcome cancer resistance. In addition, the use of nanoparticles (NPs), especially theranostic NPs permits for better monitoring of the response during treatment, and its combination with immunotherapy, promising strategies for providing a new treatment. This chapter provides an overview of these new advances in treating GC cancer.

Keywords

  • gastric cancer
  • immunotherapy
  • Theranostic
  • precision medicine
  • tumor microenvironment

1. Introduction

Gastric cancer (GC) is the fifth most prevalent cancer and the third leading cause of cancer-related death. The highest incidence of GC occurs in East Asia, followed by Eastern and Central Europe, and it is more prevalent in men. Because of the poor prognosis, many GC cases are diagnosed at advanced stages, limiting treatment options, and increasing mortality [1, 2]. Exposure to Hp, dietary risk factors, obesity, tobacco, meat, alcohol consumption, genetics, and socioeconomic status play a role in its development. More than 95% of GCs are adenocarcinomas classified by anatomic location and histologic type (Table 1) [3, 4, 5, 6, 7, 8, 9]. GC is generally treated with a combination of surgery, endoscopy, and chemotherapy, which can have side effects that can have a substantial influence on patients’ quality of life. The physical removal of malignant tissue is a key component of stomach cancer therapy. Minimally invasive procedures like laparoscopic surgery and endoscopy are used for early detection and removal of tumors. Endoscopic mucosal resection (EMR) and endoscopic submucosal dissection (ESD) are two endoscopic methods routinely used to remove early-stage stomach tumors. Despite its serious side effects, chemotherapy is still used as a treatment for advanced stomach cancer. Combination chemotherapy treatments have been found to enhance survival rates. Nausea, vomiting, exhaustion, and hair loss are some of the side effects of chemotherapy, which can be severe. On the other hand, the tumor microenvironment (TME) and multi-drug resistance by mutations in cancer cells and ABC family pumps (ATP-binding cassette) reduce its therapeutic effect. New treatment methods like immunotherapy and nanoparticles are being used to target and eliminate cancer cells. In the following sections, we will discuss all the above in detail [10, 11, 12, 13, 14].

Gastric cancer
Anatomic locationHistological type (Lauren classification)
Cardia (Proximal)Non-cardia (Distal)DiffuseIntestinal
  • Mainly in developed countries (more common in Western countries)

  • Risk factors: smoking (also in combination with drinking), consumption of food containing carcinogens, and exposure to Epstein-Barr virus (EBV)

  • less lymph-node metastasis, good differentiation, Bigger in size, poor differentiation

  • Molecular pathology: CIN and EBV type

  • Survival: Higher 5-year survival rate

  • Mainly in developing countries (more common in East Asian populations)

  • Risk factors: H. pylori infection (strong correlation), smoking, high-salt diet, food with carcinogens

  • more lymph-node metastasis, Bigger in size, poor differentiation

  • Molecular pathology: MSI type

  • Survival: Lower 5-year survival rate

  • Is correlated with non-cardia

  • prevalent in low-risk areas

  • mainly associated with heritable genetic abnormalities

  • frequently occurs in the gastric antrum

  • diffusely infiltrating the gastric wall in a desmoplastic stroma

  • characterized by poorly differentiated and discohesive tumor cells that may have either a signet-ring or non-signet-ring morphology.

  • tumor cells lack adhesion

  • Is correlated with cardio

  • more frequently in high-risk areas

  • mainly found in the cardia and gastric fundus

  • most commonly occurs in elderly male

  • is more associated with environmental factors

  • tumor cells exhibit adhesion

  • are arranged in tubular or glandular formations

  • affects the gastric antrum

Table 1.

The categorization and features of adenocarcinoma in GC.

Advertisement

2. Tumor microenvironment

The extracellular matrix that encompasses a range of cells, including cancer cells, cancer-associated fibroblasts (CAFs), pericytes, and other immune cells, is known as the TEM. The milieu around tumors, primarily made up of stromal and immunological cells, continuously affects the tumor cells [15].

2.1 Cancer-associated fibroblasts (CAFs)

About 70% of the cells in tumor tissues are CAFs, which are significant elements of malignancies such as GC [16]. They control processes such as angiogenesis, chemoresistance, and metabolic reprogramming, making them crucial in determining the spread of cancer. CAFs particularly control GC cell signaling, encouraging migration, invasion, and proliferation. They can also control immune cell activity and induce hypoxic or angiogenic circumstances through various signaling pathways. Furthermore, by secreting cytokines and chemokines, expressing cell surface receptor proteins, and remodeling the extracellular matrix (ECM), CAFs can control immune cell activity and their location and movement within the TME [17, 18, 19, 20, 21]. Subtypes of CAFs that co-expressed FAP and INHBA markers were discovered to be connected with a more advanced stage in the TME of GC. Myofibroblasts, pericytes, eCAFs, and iCAFs are the four main subpopulations of fibroblasts that were found to contribute to diffuse GC carcinogenesis. iCAFs contribute to diffuse GC carcinogenesis and may be implicated in DGC’s de novo carcinogenesis. CAFs not only shape the ECM but also supply signaling molecules that change the behavior of tumors and promote tumor survival. The Wnt/PCP signaling pathway is critical for invasion and metastasis in GC. CAFs generate WNT5A, which is the essential PCP ligand, and GC cells respond to this signal by enhancing polarized migration. CAFs secrete certain biological molecules such as Transforming Growth Factor-β (TGF-β), IL-6, and Epidermal growth factor (EGF) to promote tumor malignant phenotype, which includes tumor neovascularization and immune evasion, ultimately resulting in tumor deterioration. CAFs can also control tumor metabolism by creating an acidic milieu that inhibits the function of immune cells. Furthermore, tumor cells can exploit the metabolites of pyruvate and lactic acid generated by CAFs as nutrients to promote their proliferation [22, 23, 24, 25, 26, 27].

2.2 Immune cells

For immune editing and immunosurveillance against cancer, the host immunological response is essential. Target cells for immunotherapy include natural killer (NK) cells, dendritic cells (DCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), T cells, B cells, and regulatory T cells (Tregs). These cells are found in the TEM [28, 29, 30]. Through a variety of tumor cells can avoid immune system recognition and elimination. M2 TAMs, N2 TANs, MDSCs, regulatory B cells (Bregs), effector Tregs (eTregs), and inhibitory targets on different immune cells are among them that are important in tumor immune escape while tumor-infiltrating DCs, NK, M1 TAMs, and N1 TANs are helpful in anti-tumor immunity (Figure 1) [30].

Figure 1.

Immune cell-targeted treatment can be accomplished by either boosting anti-tumor immune cell counts or functions or by decreasing immunosuppressive cell counts or functions [30]. Copyright 2022 Frontiersin.

2.2.1 Dendritic cells (DCs)

Dendritic cells play a crucial role in activating T cells and bridging innate and adaptive immunity. Maintaining a sufficient density of mature DCs within tumors is essential for prolonged life of patients with advanced GC. There are two types of DCs: conventional DCs (cDCs; sometimes called myeloid DCs) and plasmacytoid DCs (pDCs) pDCs have gained interest in recent years for their ability to present antigens, secrete cytokines, and promote the proliferation and functionality of immune cells [31, 32].

2.2.2 Natural killer cells

NK cells play a crucial role in regulating the immune system against cancer cells and intracellular viruses. They identify tumor cells via NKG2D and eliminate them using perforin, death receptor signaling, or granzymes that generate cytokines and chemokines [33, 34, 35]. NK cells contain inhibitory receptors on their surface, which allows tumor cells to connect to them and evade NK cell killing. On the other hand, excess TGF-β, together with other chemokines and anti-inflammatory cytokines, can prevent NK cell activation in the TEM; produce some immunosuppressive factors, and downregulate NK cell activating receptors such as NKp30, NKp44, NKG2D, and CD16 as well as co-receptors including NKp80 and DNAM-1. It also upregulates checkpoint receptors such as TIGIT, TIM-3, LAG-3, and PD-1 [36, 37, 38].

2.2.3 Tumor-associated macrophages

Macrophages are a type of immune cell that infiltrate solid tumors. They are divided into two subgroups: M1 and M2. M1 macrophages have proinflammatory effects and produce High amounts of TNF-α, IFN-γ, IL-1β, IL-6, IL-12, CXCL9, and CXCL10 that stimulate type 1 (polarizing and recruiting Th1) responses aiding in the elimination of infections and tumor cells. On the other hand, M2 macrophages have anti-inflammatory effects and produce TGF-β1 (TGF-β1), IL-4, and IL-10 that have tumorigenic qualities [39, 40, 41, 42]. Macrophages play a crucial role in gastric inflammation during H. pylori infection. In GC tissue, macrophages are more abundant and their gene expression patterns are diverse. Macrophage-GC cell crosstalk contributes to the development of an immunosuppressive milieu and the progression of GC. M2 features are linked to a poor prognosis in many tumor types [43, 44, 45, 46, 47].

2.2.4 Tumor-associated neutrophils

As the initial line of defense against infection and inflammation, neutrophils play a crucial role in Innate immune responses. Neutrophils are associated with a worse prognosis in GC patients and are abundant in GC tissue [48, 49, 50]. Neutrophils are one kind of immune cell that promotes angiogenesis and inhibits the anti-tumor immune response, which contributes to the development of tumors [32]. GC-derived GM-CSF suppresses T-cell activity to advance GC by activating neutrophils and eliciting PD-L1 expression via the JK-STAT3 signaling pathway. Additionally, TANs secrete cytokines that aid in GC invasion and migration, including IL-1β, IL-6, IL-8, IL-17a, and IL-23 [48, 51, 52].

2.2.5 Myeloid-derived suppressor cells

The primary role of myeloid-derived cells, or MDSCs, is immunosuppression. They are a very diverse collection of cells. Malignant tumors with MDSC growth had a poor prognosis and were linked to treatment resistance. Increased MDSC levels were linked to later tumor stages, a worse prognosis, increased mortality, and a higher chance of tumor progression and recurrence in GC patient’s survival [53, 54, 55]. These relationships were also seen between MDSC levels and the cancer stage. MDSCs are involved in the production of immunosuppressive TME as well as the advancement and metastasis of tumors (Figure 2) [57, 58, 59].

Figure 2.

The processes via which gastrointestinal (GI) cancer is immunosuppressed by MDSC. MDSCs block the activity of DCs, prevent DCs from presenting antigens to CD4+ T cells, repress the growth and function of NK and T cells, decrease CD8+ T-cell infiltration, support M2 macrophage development, and stimulate the expansion and immunosuppression of Tregs. Moreover, nitric oxide (NO) generation and TGF-β-induced suppression of NKG2D have an impact on ADCC function and NK cell anergy, respectively. To encourage the proliferation and metastasis of GI cancer cells, MDSCs produce exosomes, matrix metalloproteinases (MMPs), and VEGF [56]. Copyright 2021 Wiley.

2.2.6 B cells

In addition to acting as APCs, B cells also create and ingest cytokines including IL-6, IFN-γ, and TNF-a, which help to develop CD4+ and memory T cells [60]. Research has demonstrated that effector T cells, NK cells, and TAMs, as well as T cells and other immune cells, are the primary mediators of antitumoral activity, which is inhibited by a subset of B regulatory cells called Bregs [61].

2.2.7 T cells

T lymphocytes come in different varieties: helper T cells (CD4+ cells), Tregs, and cytotoxic T cells (CD8+ cells). The TME’s CD8+ T cell subset is widely regarded as having immunological activation to produce antitumor effects, and the prognosis and treatment results of GC malignancies are strongly correlated with the subsets’ variety and density. Tumor prognoses vary depending on different T cell subsets. T-cell subsets that are advantageous to the prognosis and survival of GC include those with a high T helper (TH)1/TH2 ratio, high CD45RO memory T cells, and high levels of infiltration of CD8+ and CD4+ T cells [62, 63, 64].

2.2.8 Regulatory T cells

From 5–10% of peripheral CD4+ T cells are Tregs, which are responsible for maintaining immunological tolerance linked to tumor immunity. Numerous investigations have documented the significance of Tregs as a constituent of immune-suppressive cells, which expedite the growth of tumors in diverse malignancies by inhibiting T cell proliferation, antigen presentation, and cytokine generation. Based on how FOXP3 and CD45RA are expressed, TME Tregs may be divided into three groups: effector Tregs (eTregs) (FOXP3highCD45RA), naïve Tregs (FOXP3lowCD45RA+), and non-Tregs (FOXP3lowCD45RA). Non-Tregs can release pro-inflammatory cytokines but are unable to have an inhibitory impact. The function of eTregs is persistent and has substantial suppressive activity, while naïve Tregs only exhibit mild suppression [65, 66, 67]. In Figure 3, the various T cells, B cells, and Tregs are discussed concerning the GC microenvironment; Figure 4 also illustrates the significance of TAMs, NK cells, DC cells, and neutrophils in this context.

Figure 3.

The functions of various B cells, Tregs, and T cells in the stomach cancer microenvironment. In GC, certain Tregs and dysfunctional CTLs accelerate disease progression, while specific T cell subsets indicate poor outcomes. Conversely, CXCR5+ CD8+ T cells and CD103+ Trm cells are associated with better survival. PMN-MDSCs contribute to immunosuppression as cancer develops. Elevated CD73 expression in tumors enhances CD8+ T cell infiltration. Reduced expression of homing receptors in Aim2−/− mice leads to increased gastric CD8+ T cell frequency and metaplasia. Higher levels of NKG2D correlate with improved survival, as it interacts with tumor cells’ MICA/B and ULBPs. CD155 on tumor cells inhibits TIGIT+ CD8 T cells’ glucose uptake, diminishing their function. ICOS+ CD4+ T cells hinder CD8+ T cell proliferation, hastening disease advancement [29]. Copyright 2023 Springer.

Figure 4.

The functions of DC cells, neutrophils, NK cells, and TAMs in the GC microenvironment. In gastric cancer microenvironment, TAMs release immunosuppressive chemicals like CH13HL1, CXCL8, and CCL5, promoting tumor growth. Neutrophils’ secretion of MMP-9 and IL-17 correlates with poor prognosis. IL-33-driven mast cell buildup, facilitated by macrophage mobilization, contributes to GC progression in certain animal models. High levels of CD68+, CD163+, and IL10+ macrophages correlate with decreased survival rates. DC-SIGN+ macrophages elevate FOXP3 Treg levels, associated with lower overall survival. TAMs reduce NK cell percentage and function, while NK cells express CD16 and exhibit direct antitumor effects. Poor prognosis in GC patients is linked to high CD11b levels in DC cells, which lowers MHC class II expression. Neutrophils, driven by GC cell-derived exosomes and GM-CSF, promote tumor migration and enhance their own survival [29]. Copyright 2023 Springer.

We have summarized the findings of various studies to enhance understanding regarding the role of immune cells, such as T cells, TAM cells, B cells, NK cells, DC cells, neutrophils, and cytokines secreted by stromal cells in GC progression. You can find this summary in Table 2 for your better understanding.

Immune cellsCancer typeFunctionsRef.
Dendritic cells
DC cellsGCforeseen prognosis is unsatisfactory[68]
Plasmacytoid DC cellsGCImproved the efficiency of can-
cer treatments
[31]
Natural killer cells
NK cellsGCForesaw a more satisfactory 5-year survival rate compared to lysis activity of less than 25%[69]
NK cellsGCEffectively, trastuzumab-targeted tumor cells with a potent killing[70]
NK cellsGCCompared to low NK cell density has a more favorable prognosis GC[71]
NK cellsGCSurvival is predicted to be longer[72]
NK cellsGCTumor cells exhibit limited immune evasion.[73]
NK cellsGCPredicted better OS[74]
High-affinity NK cellsGCInduced direct antitumor effects[75]
Tumor-associated macrophages
TAMsGCPrompted HMGB1 expression contributes to chemoresistance to 5-Fu[76]
TAMsGCBoosted PD-L1 expression and
tumor metastasis
[77]
TAMsGCImpaired NK cell proportion
and function
[78]
TAMsGCEnhanced DNMT1 and low GSN expression to promote tumor growth[79]
DC-SIGN+ TAMsGCIndicated poor OS and resistance to ACT such as 5-Fu[80]
CD68+ CD163+ not MARCO+ TAMsGastroesophageal adenocarcinomaIndicted poor survival[81]
IL10+ TAMsGCPromoted regulatory T cell infiltration and CD8+ T cell dysfunction[82]
Neutrophils
NeutrophilsGCInduced tumor migration[83]
NeutrophilsGCExecuted Immunosuppression[49]
Neutrophils in the peripheral
blood
GCIndicated poor prognosis[84]
Myeloid-derived Suppressor cells
PMN-MDSCsGCExecuted immunosuppressive
function
[85]
T cells
CD8+ T cellsGastric and gastroesophageal junction (G/GEJ) adenocarcinomasShortened PFS and OS[86]
CD8+ T cellsGC with high CD73 expressionThe phenotype of the CD8+ T cells indicated that they were dysfunctional.[87]
CXCR5 + CD8+ T cellsGC with TNM II + III
stage
Benefited from ACT[88]
CXCL13+ CD8+ T cellsGCIndicated poor prognosis[89]
(ICOS+) CD4+ T cellsGCImpeded the proliferation of CD8+ T cells[90]
Foxp3+ RORγt+ T cellsGCPredicted poor overall survival[91]
Tumor-infiltrating γδT cellsGC with TNM II + III
stage
Benefited from 5-Fu ACT[92]
CD69+ CD103+ TrmGastric adenocarcinomaPredicted better survival[93]
HLA-1-preserved type/PDL1+Microsatellite-unstable GC
tumors
Indicated good prognosis[94]
TIM3+ cellsGCHad poorer OS and disease-free
survival (DFS)
[95]
Regulatory T Cells
TNFR2+ TregsGCPromoted the GC progression[96]
CD4 + CD25 + FOXP3+ TregsGCHelped immune-suppressive roles of Tregs[97]
CD45RA − CCR7− TregsGCExerted immunosuppression[98]
CD4+ FOXP3+ CD25high TregsGCExerted Immunosuppression[99]
FOXP3+ TregsStage I–II GC patientsForeseen more acceptable prognosis[100]
B cells
B regulatory cellsGCInhibited antitumoral activity[61]
CD20+ B cellsGCProspered disease-free survival and overall survival rate[101]
Cytokines or chemokinesCancer typeFunctionsRef
CCL28GCDrove GC tumor progression[102]
IL-9GCAchieved additional benefits from 5-Fu-based adjuvant chemotherapy and Lengthy OS[103]
IL-15GCPromoted tumor progression[104]
IL-17GCBenefited from adjuvant chemo-
therapy
[105]
IL17BGCAccelerated cancer stem cell tumorigenesis and self-renewal[106]

Table 2.

A review of the immune system’s role in GC disease progression, by different immune cells, cytokines, and chemokines.

Advertisement

3. Resistance to the treatment of gastric cancer

GC is known for its resistance to standard treatments like chemotherapy and radiotherapy. Immunotherapy is a promising approach, but its effectiveness is often hindered by TEM of GC. Surgical resection with lymphadenectomy is the primary treatment strategy for GC. Surgery is the primary option for GC due to its resistance to chemotherapy and radiation [107, 108]. The TEM in GC is distinct from normal tissue, leading to fluctuations in glucose, lactate, oxygen tension, and acidic pH. Glycolysis leads to lactic acid production and subsequent tumor acidosis. Aggressive tumors rely heavily on glycolysis for energy production, even in the presence of adequate oxygen, exacerbating the acidic environment [109, 110].

3.1 Hypoxia

GC microenvironment is characterized notably by hypoxia and inflammation. Hypoxia, defined as a reduction in normal tissue oxygen levels, emerges due to rapid tumor cell proliferation surpassing the oxygen delivery capabilities of existing blood vessels. This condition triggers a series of cellular responses in tumor cells, enabling them to adapt and thrive in this low-oxygen environment. These cells undergo a range of molecular and physiological adaptations under hypoxic conditions, which often include upregulation of hypoxia-inducible factors (HIFs), alterations in metabolic pathways such as a shift towards anaerobic glycolysis, and the activation of survival and proliferative signaling pathways [111, 112, 113]. hypoxia in TEM is linked to poor clinical outcomes and treatment resistance in GC. Hypoxia triggers hypoxia-inducible factor-1 (HIF-1), which regulates the expression of many cancer-promoting molecules. HIF-1 is crucial in promoting angiogenesis, which is vital for tumor growth and spread in hypoxic conditions. Hypoxia also triggers the production of immune checkpoint molecules like PD-L1, which naturally slow down T-cell activity. Overexpression of PD-L1 hinders the effectiveness of immunotherapies, like checkpoint inhibitors, that aim to activate the immune system against cancer cells. Hypoxic GC cells show higher levels of PD-L1 expression, which contributes to the evasion of immune surveillance [30, 114, 115, 116].

3.2 Oxidative stress

Oxidative stress affects the resistance of GC to immunotherapy. In an oxidative stress state is a lack of balance between the body’s antioxidant defenses and the production of reactive oxygen species (ROS). High levels of ROS in GC can lead to genetic mutations, cancer cell proliferation, and metastasis, which often result in an increased resistance to cancer therapy, including immunotherapy [117, 118, 119]. For instance, oxidative stress can impair the effectiveness of T cells, a crucial part of the immune response in immunotherapy. However, targeting oxidative stress in treating GC is challenging due to the complex role of ROS in cancer biology, where it can both promote and inhibit tumor growth. It also requires sophisticated techniques and a deep understanding of tumor physiology to accurately assess and modulate oxidative stress levels within tumors [120, 121].

3.3 Acidosis

Changes in metabolic processes, specifically the Warburg effect, cause acidosis in tumors. The cancer cells consume glucose through glycolysis instead of oxidative phosphorylation, which results in the production of lactic acid. This, in turn, lowers the pH of the environment surrounding the tumor [122, 123]. This can impair the effectiveness of immunotherapies by inhibiting the function and survival of T cells and upregulating immune checkpoint molecules. The acidic environment also inhibits the effector functions of activated CD8+ T-cells. The study of Wu shows that acidic paracortical zones in lymph nodes inhibit the effector functions of activated CD8+ T-cells and hinder T-cell glycolysis. This low pH, is a result of the acid inhibition of monocarboxylate transporters (MCTs), leading to a decrease in the glycolytic rate [124]. Despite this, the acidic environment does not prevent the initial activation of native T-cells by DCs. Low pH conditions reduce cytokine production and proliferative capacity of T cells, which are crucial for their cancer-fighting abilities [125, 126]. Acidosis can also upregulate PD-L1 expression, contributing to the resistance to immunotherapy [127, 128].

Advertisement

4. New approaches in cancer immunotherapy

Despite advancements in GC treatment, there’s debate on the most effective approach. Multimodal treatments have improved patient survival, but over 70% of patients cannot be rescued. The side effects of therapy can lower the quality of life, which can impact the outcome. This fact has made researchers explore alternative approaches to managing GC [129, 130]. For GC treatment, there are several therapeutic methods with a different level of invasiveness from surgery to adjuvant therapies yet surgery is considered the priority therapy. Although, whether preoperative or postoperative, has shown improvement in the overall outcome of GC treatment, each of the conventional therapies comes with a spectrum of complications, ranging from severe to mild. Despite the radiotherapy, chemotherapy, and chemoradiotherapy have advanced, surgical resection remains the primary treatment option for GC. However, it is a high-risk procedure with significant morbidity and mortality, especially in elderly patients, has a lower tolerance for anesthesia and surgery due to a decline in organ function and an increase in internal diseases. While postoperative mortality has decreased by more than 50% in four decades, significant surgical morbidity following gastrectomy continues to pose challenges [131, 132]. Chemotherapy has been found to have comparable or better survival rates compared to surgeries for GC. Palliative chemotherapy enhances survival rates without impacting the quality of life significantly. Nausea, vomiting, and diarrhea are some common side effects However, acute chemotherapy complications like GI hemorrhage, perforation, nausea, vomiting, diarrhea, and typhlitis, from neutropenia by chemotherapy manifest as localized inflammation in the caecal wall [133, 134, 135]. Other approaches, such as radiation therapy, have been investigated to improve outcomes. However, radiation therapy can have severe side effects such as skin problems, diarrhea, fatigue, and low blood cell counts. Patients may encounter GI side effects [136, 137].

4.1 Novel immunotherapy methods

Immunotherapy emerged as a potent clinical approach to cancer therapy. The quantity of approved immunotherapy drugs has been steadily rising and there are also numerous treatments undergoing preclinical development and clinical trials. Immunotherapies for GI cancers include a range of approaches such as checkpoint inhibitors, cancer vaccines, adaptive cell transfer, and cytokines [138, 139, 140]. There are also complications with conventional immunotherapy methods such as Colitis secondary to immunotherapy coupled with diarrhea, abdominal pain, nausea, and vomiting. However, research has led to advancements in immunotherapy and other novel approaches to address these limitations. Subsequently, novel approaches to GC treatment are also being discussed [141, 142, 143, 144].

4.1.1 Immune checkpoint inhibitors (ICIs)

T cells combat cancer by recognizing antigenic peptides presented on the Human Leukocyte Antigen (HLA) through T-cell receptor (TCR). This process is called immune surveillance and prevents tumor development. TSA is a specific target antigen for TCR on tumor cells. When the immune system becomes “blind” to TSA, tumors can grow uncontrollably, a phenomenon known as “tumor escape”. Immune checkpoints serve as intrinsic components of the immune system, responsible for moderating the strength of immune responses to prevent harm to healthy cells. They come into play when proteins on T cells’ surface, recognize and interact with partner proteins on other cells, including certain tumor cells. These partner proteins, known as immune checkpoint proteins, trigger an “off” signal upon binding with the checkpoint proteins, thereby dampening the immune response and preventing the destruction of cancer cells [139]. Immunotherapy drugs known as immune checkpoint inhibitors function by obstructing the binding of checkpoint proteins with their partners, thus thwarting the transmission of the off signal. Consequently, this enables T cells to target and eliminate cancer cells more effectively. Upon encountering cancer cells, cytotoxic T lymphocytes (CTLs) release interferon (IFN)-γ. Following its release, IFN-γ binds to IFN-γ receptors (IFN-γR) present on the surface of tumor cells. This interaction triggers the activation of the JAK-STAT signaling pathway, resulting in the increased expression of programmed death ligand-1 (PD-L1). Subsequently, when PD-1 receptors are blocked, inhibitory signals are transmitted to T cells, impairing their capacity to eradicate tumor cells (Figure 5) [146, 147]. PD-1 is significantly upregulated in thymocytes, NK cells, myeloid dendritic cells, and B and T cells. During inflammatory conditions, PD-L1 and 2 form functional interactions with PD-1 to protect normal tissues. However, this mechanism simultaneously enables tumors to evade immune detection. Therefore, anti-PD-1 agents showed the potential to restore T-cell activation by disrupting this interaction. mAbs targeting ICIs, like PD-1 and its corresponding ligand, have shown significant improvements in extending survival rates across different types of cancers, including advanced GC [148]. Nivolumab and Pembrolizumab are human mAbs of the IgG4 class, specifically engineered to block the PD-1 pathway (See Table 3). CTLA-4 is another critical immune checkpoint molecule found on the surface of T cells. This molecule interacts closely with CD28 and serves a crucial role in downregulating T-cell activation. The CTLA-4 pathway acts in an inhibitory role similar to that of PD-1, when bound by B7–1/2 on an APC [140]. This prevents T cells from binding to the CD28 receptor which is a co-stimulatory molecule on the surface of CD4 cells [151]. Thereby it facilitates the immune escape of tumors.

Figure 5.

Targets and immune checkpoints for GC [145]. Copyright 2021 Elsevier.

StudyTreatment line/PhasePrimary endpointRegimenDCR/ ORR/ (%)
KEYNOTE-059≥ 3rd line/ IIOS and PFS in PD-L1CPS ≥ 1Pembrolizumab−/57.1
KEYNOTE-0612nd line/ IIIOS and PFS in PD-L1CPS ≥ 1Pembrolizumab−/46.7
KEYNOTE-0621st line/ IIIOS and PFS in PD-L1CPS ≥ 1CT + Pembrolizumab−/64.7
KEYNOTE-164≥ 2nd line/ IIORRPembrolizumab57/33
KEYNOTE-1771st line/ IIIPFS and OS200 mg every 3 weeks64.7/43.8
CheckMate-1421st line/ IIORRCT84/69
CheckMate-6491st line/ IIIPS and PFS in PD-L1 CPS ≥ 5Nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks−/70

Table 3.

Summarized overview of ongoing trials of checkpoint inhibitor drugs [149, 150].

DCR (disease control rate); PFS (progression-free survival); ORR (objective response rate); CT (chemotherapy).

4.1.1.1 CTLA-4 inhibitors

Antibodies targeting CTLA-4 bind specifically to these small molecules and alleviate T-cell suppression. Key examples approved of anti-CTLA-4 antibodies are Tremelimumab and ipilimumab (Table 3). Ipilimumab obtained FDA approval in 2011 for the management of advanced melanoma. In a clinical study, Ipilimumab showed an objective response rate of 14% in patients with advanced gastric cancer. However, in another trial, Ipilimumab did not increase survival rates when employed as maintenance therapy following first-line chemotherapy for advanced GC or gastroesophageal junction cancer. T Tremelimumab, which acts as a specific CTLA-4 inhibitor is an IgG2 monoclonal human antibody that augments T-cell functionality. In a trial, Tremelimumab administered as a subsequent therapeutic approach showed a median PFS of 1.7 months and a median OS of 7.7 months in patients diagnosed with gastric cancer or gastroesophageal junction cancer following chemotherapy [145, 152, 153].

4.1.1.2 PD-1/PD-L1 inhibitors

Pembrolizumab and Nivolumab have been studied as potential treatments for advanced gastric cancer. In 2017, pembrolizumab was given approval by the Food and Drug Administration (FDA) as a third-line treatment for patients with positive CPS for PD-L1. Nivolumab showed significant survival rate enhancement and received approval as a third-line treatment for GC in Japan. Avelumab did not show significant improvement in overall survival when used as a maintenance and monotherapy treatment following first-line chemotherapy for advanced GC or gastric junction cancer. Overall, PD-1/PD-L1 inhibitors demonstrate clinical effectiveness in the management of advanced GC or gastroesophageal junction cancer., However, the benefits of monotherapy are relatively modest, particularly in later lines of treatment. Despite the approval of anti-PD-1 and PD-L1 therapies as monotherapies and in later lines of treatment, it is not likely that they will emerge as first-line therapies for GC [145, 154, 155].

4.1.1.3 Combinational therapy of Anti CTLA-4 and PD-1

The combination of anti-CTLA-4 and anti-PD-1 antibodies led to the inhibition of epithelial-mesenchymal transition (EMT) and reduced migration and invasion; a notable suppression of cell proliferation and apoptosis induction in MGC-803 and MKN-45 cells. Results from western blot analysis exhibited that this combination reduced the activation of β-catenin, MAPK, and PI3K/AKT signaling pathways. Additionally, in vivo, studies demonstrated that the combination therapy effectively inhibited tumor formation. Furthermore, transfection with si-PD-1 and si-CTLA resulted in significantly decreased levels of CTLA-4 and PD-1 in transcript levels. These findings suggest promising outcomes for the mentioned combination therapy in GC, yet further investigations are required (Table 4) [160].

Drugs(mAb)Target therapiesVEGFR-2Ramucirumab
Apatinib
Regorafenib
Lenvatinib
HER-2Trastuzumab
Margetuximab
ICIsPD-1Camrelizumab (SHR-1210)
Sintilimab
Nivolumab
Pembrolizumab
Toripalimab (ISO01)
Tislelizumab
PD-L1Avelumab
Atezolizumab
Durvalumab
Sugemalimab (CS1001)
CTLA-4Tremelimumab
Ipilimumab

Table 4.

mAb for targeted treatment of GC by inhibiting cell surface receptors [145, 156, 157, 158, 159].

4.1.2 Cellular immunotherapy

The utilization of immune cells as therapeutic interventions for cancer is referred to as cellular immunotherapy. T lymphocytes and NK cells cultivated in vitro are pivotal elements of these therapeutic strategies. These cells can undergo genetic modification to express certain T cell receptors (TCR-T immunotherapy) or chimeric antigen receptors (CAR-T Cell therapy) against desired targets [145].

4.1.2.1 NK cell transfer therapy

NK cells indeed hold a pivotal role in combating tumor cells by hindering the inception and advancement of tumors (Section 1.2.2) [30]. They induce cytotoxicity by release of granzyme and perforin, antibody-dependent cytotoxicity (ADCC), secretion of TNF-α and IFN-γ, and induction of apoptosis through FAS/FASL and TRAIL/TRAILR complex interactions and modulate immune response. However, malignant cells develop mechanisms to evade immune responses. Moreover, NK cells’ quantity and functionality decline as GC advances. Consequently, rectifying NK cell dysfunction may present a promising therapeutic avenue for GC. NK cell adoptive therapy presents a promising therapeutic avenue for gastric cancer. A Phase I clinical trial validated the safety and tolerability of autologous expanded NK cell therapy, which indicates the potential for effective combination treatment with other agents [161, 162].

4.1.2.2 CAR-T cell therapies

Targeted T-cell immunotherapy is the most thoroughly investigated and promising therapeutic pathway in the realm of GC. They have shown impressive results in clinical studies, particularly within hematological malignancies. These cells are genetically engineered to attack cancerous cells. A specific single-chain variable fragment (scFv) binding to a cancerous cell target is essential to engineering CAR-T cells. T cells are subsequently genetically engineered to express a chimeric receptor specifically designed to recognize and attack tumor cells (Figure 5) [163, 164]. Claudin18.2 is a membrane protein specific to the stomach. This molecule has been identified as a promising target in therapeutic approaches in GC as well as other types of cancer. CAR-T cells targeted towards this molecule eradicated gastric tumors in mice without causing unspecific toxicity. The latest clinical findings on GC and pancreatic cancer utilizing CAR-Claudin18.2 T cells have shown promise with a 33.3% overall objective response rate [165, 166]. NK group 2, member D (NKG2D) ligand AKA stress-induced ligand (NKG2DL) is a potential target for GC therapies. This molecule is typically expressed in GC cell lines. NKG2D-CAR-T cells, generated through the modification of CAR-T cells using second-generation CAR vectors, exhibit remarkable anti-tumor efficacy in both animal models and laboratory investigations [167]. HER2 molecule is an abnormally over-expressed molecule in 10–20% of GCs. HER2-CAR-T cells have demonstrated significant and persistent anti-tumor effects against GC cells in laboratory and animal models [168, 169]. Folate receptor 1 (FOLR1) serves as a target molecule that is prominently found overexpressed on the cell surface in more than 30% of GC patients, However, its expression in normal tissue is uncommon. This unique characteristic positions it as a promising target for CAR-T immunotherapy [170]. Prostate stem cell antigen (PSCA), is recognized for its glycosylphosphatidylinositol (GPI) anchored cell surface protein nature and presents as another target for CAR-T cell therapy in GC [171]. Mesothelin (MSLN) has also been considered a cellular target for CAR-T therapy in treating GC. Third-generation CAR-T cells (M28z10T) developed to specifically target MSLN, showed potent anti-tumor activity, underscoring their potential as a promising therapeutic strategy for GC [172].

4.1.2.3 TCR-T immunotherapy

This strategy involves the insertion of TCR genes targeting tumor antigens into the circulating T cells of patients, representing a modified form of adoptive T cell-based cancer immunotherapy. Encouraging outcomes have been observed with this approach across a spectrum of solid tumor types, including multiple myeloma, lung cancer, and synovial sarcoma [173, 174, 175]. Kita-Kyushu Lung Cancer Antigen-1 (KK-LC-1) is frequently identified as a cancer germline antigen in various epithelial cancers, including non-small cell lung cancer, triple-negative breast cancer, cervical cancer, and GC [176, 177]. CT83 expression is remarkably high in GC, and tumor lines positive for CT83 may be recognized by genetically modified T cells with the KK-LC-1 TCR (KK-LC-1 TCR-Ts) in vitro [178]. Another target for TCR-T therapy is potentially the New York esophageal squamous cell carcinoma 1 (NY-ESO-1). Its discovery is rooted in its ability to elicit detectable antibody responses in cancer patients. It is notable that following surgery, those without relapses had sustained reductions in or eradication of the serum levels of NY-ESO-1 antibodies [179]. These results demonstrated the KK-LC-1 TCR-Ts’ encouraging potential for treating GC.

4.1.3 Vaccine treatment

Cancer therapeutic vaccines fall under the umbrella of active immunotherapy, designed to provoke a focused immune reaction against tumor antigens [180]. Peptides, DC, autologous tumor cells, and genetically engineered vaccines are among the vaccines used against cancer. A study evaluated the therapeutic potential of combining adjuvant immunotherapy with chemotherapy in GC patients, employing autologous tumor-derived Gp96 immunization considering that tumor antigens were the initial focus of tumor vaccination research [181]. In the group receiving the vaccine, patients exhibited enhanced rates of disease-free survival and 2-year OS compared to those undergoing chemotherapy alone, standing at 81.9% versus 67.9%. Another investigation, which focused on 9 individuals with advanced GC treated using the HER2/DC vaccine, documented partial clinical remission in a single patient, coupled with a decline in tumor marker levels such as CEA and CA19–9. Furthermore, there were observable anti-tumor effects observed in patients with advanced GC administered Melanoma-associated antigen A3 peptide (MAGE-A3)/DC [182].

Reportedly, 85% of diagnosed GC patients exhibit expression of Lymphocyte antigen 6 complex locus K (LY6K). In six patients with advanced GC, a Phase I clinical trial that investigated the vaccinations with LY6K-derived peptide showed that three out of 6 had more stable disease states and one had a 20% reduction in the size of metastases in their liver [183].

Advertisement

5. Nanomedicine and targeted therapies

Nanomedicine is a crucial tool in cancer management. Nanoparticles (NPs) can enhance the effectiveness of anticancer agents or function as anticancer agents themselves, targeting tumor cells in both active and inactive states based on the tumor type and TME conditions. This overcomes traditional cancer therapy limitations [184].

Polymeric NPs are used as carriers for chemotherapeutic drugs, due to their unique structures that encapsulate drug molecules. They address the limitations associated with these drugs such as instability, low solubility, toxicity, and poor permeability, improving cancer treatment outcomes [185, 186]. Numerous studies have explored the use of polymeric NPs (NPs) loaded with chemotherapeutic drugs like paclitaxel (PCT), doxorubicin (DOX), docetaxel (DCT), 5-fluorouracil (5FU), and other drugs for the treatment of GC. Recently, there has been increasing interest in hybrid drug delivery, which involves delivering two or more chemotherapeutic agents simultaneously, despite their delivery to the tumor site. This approach holds promise as a potent strategy to enhance GC treatment efficacy. However, the most significant challenge remains to achieve a more precise and targeted delivery of drug agents to the tumors’ site, even with NPs. This difficulty arises from the varied physicochemical properties of anticancer agents. For example, researchers have developed NPs based on polyethylene glycol and poly(lactide-co-glycolide) (PEG-PLGA), loaded with 5-fluorouracil (SN-38-5FU@NPs) and irinotecan (SN-38), using the nanoprecipitation technique to improve the effectiveness of GC treatment [187, 188, 189]. Dendritic polymers like Poly(amidoamine) have potential for biomedical applications. PEGylated dendrimers with celastrol are used in GC therapy [184, 190]. Exosomes are nanoscale vesicles that range from 30 to 150 nanometers in diameter. They are secreted by various cell types and hold significant promise as GC therapy. Recent studies shed light on the role and outcome of exosome application in treating GC cell lines. Exosomes have potential as biomarkers for GC diagnosis and as targets for therapeutic approaches [191, 192]. Liposomes are colloidal vesicles made of phospholipids that mimic natural cell membranes. They have a high drug-loading capacity and can deliver hydrophilic and hydrophobic compounds. Liposomes are biodegradable, biocompatible, non-immunogenic, and non-toxic. They are widely used for delivering anticancer drugs in GC treatment [193, 194].

Metallic NPs are used as novel anticancer agents in the treatment of GC. They are produced by capping and reducing metal precursors like zinc acetate dihydrate, copper nitrate, gold halides, and silver nitrate [195]. Metallic NPs can be used alone or with other drugs to achieve better anticancer effects against GC. Silver, gold, nickel oxide, zinc oxide, and cobalt oxide NPs have shown outstanding outcomes against GC cells by augmenting targeting precision, facilitating controlled drug release, supporting imaging modalities, and enhancing gene modification mechanisms [196]. Gold NPs have shown potential as a versatile tool for both diagnosing and treating cancer. Studies have found them to be effective against GC cells while demonstrating good biocompatibility, In AGS cell lines, inhibiting autophagy-related pathways and up-regulating apoptotic signaling [197, 198]. Silver NPs synthesized using eco-friendly methods and natural sources have been shown to significantly inhibit the proliferation of gastric cancer (GC) cells, including the AGS and MNK45 cell lines. This inhibition is achieved by inducing apoptosis, which suggests a promising approach for effective GC therapy [184]. Additionally, nickel oxide and cobalt NPs synthesized through chemical methods displayed cytotoxicity against AGS-GC cell lines in a dose-dependent manner. Nevertheless, nickel oxide and cobalt NPs both exhibited minimal toxicity towards normal fibroblast cells (L929) [197, 198].

5.1 Theranostic NPs

Theranostic NPs are sophisticated nanosystems, that facilitate highly accurate and individualized disease management by amalgamating diagnostic and therapeutic functionality (Figure 6) [200]. Superparamagnetic iron oxide nanoparticles (SPIONs) can be used for magnetic resonance imaging (MRI), while gold nanoparticles (NPs) are well-suited for computed tomography (CT). Therefore, they are commonly used in the construction of theranostic systems. Additionally, some photosensitizers, such as chlorin e6 and IR780, have both tumor-toxic and imaging abilities. This is because they can absorb near-infrared (NIR) light to induce fluorescence and generate reactive oxygen species (ROS) and/or heat [201]. though the structure and assembly of these nano-based theranostic systems can be complex, many have already been reported, demonstrating their potential in treating gastric cancer (refer to Table 5) [200, 214].

Figure 6.

The diagram depicts NPs designed for gastric cancer theranostics. Specifically, it showcases a tumor-targeted and matrix metalloprotease-2 (MMP-2)-activatable nanoprobe (T-MAN). This nanoprobe is developed by modifying Gd-doped CuS micellar NPs with cRGD, covalently. This modification allows for selective entry and accumulation of the nanoprobe in gastric tumors via αvβ3 integrin-mediated active delivery [199]. Copyright 2019 American Chemical Society.

NanoparticlesTheranosticRef.
Therapeutic strategyImaging strategy
ICG-loaded lactosomePTTFluorescence[202]
Carbon–gold hybrid NPsPTTFluorescence[203]
RGD-CuS-Cy5.5 NPsPTTCT/MRI[204, 205]
W18O49 NPsPTTCT/fluorescence[204, 206]
PEGPCL-IR780-MET NPsPTTPA/fluorescence[206, 207]
Hyaluronidase-sensitive mesoporous silica NPsPDTFluorescence[207, 208]
Ternary copper-based chalcogenide nano platform CuS–NiS2 nanomaterialsPTT/PDTMRI[209]
Folic acid–sericin–cholesterol/IR780 micellesPTT/PDTFluorescence[201, 210]
DOX-IR820 NPsPTT/chemotherapyFluorescence[211]
IR820/paclitaxel/imiquimod/encapsulated thermosensitive liposomePTT/PDT/chemotherapyFluorescence[201, 212]
PTX-R837-IR820@TSLPTT/PDT/chemotherapy/ ImmunotherapyFluorescence[207]
Cisplatin/ICG loaded PLGA-(DSPE-PEG2000) NPsChemotherapyFluorescence[212, 213]
Chlorin e6 functionalized silk fibroin NPsChemotherapyFluorescence[208, 210]
Oxaliplatin-Au-Fe3O4-Herceptin NPsChemotherapyMRI[205, 213]

Table 5.

Nanotechnology-based theranostic(therapeutic/diagnostic) agents for GC.

Photothermal therapy (PTT); Photodynamic therapy (PDT); Arginine-Glycine-Aspartic (RGD).

Xin Luo and et.al developed a nanocarrier system that enhances siRNA delivery using a PD-L1 knock-down system. The system involved creating disulfide-polyethylene glycol-folic acid-conjugated polyethyleneimine coupled with Fe3O4 SPIONs. This approach entailed encapsulating FA-PEG-SS-PEI and SPIONs utilizing a ligand-exchange method, followed by combining this combination with cationic micelles complexed with synthesized siRNA. The product underwent characterization and exhibited excellent contrast for T2-weighted cancer MRI using cellular MRI. PD-L1 siRNAs exhibited notable knock-down of PD-L1, supporting their use as a theranostic approach [215, 216].

Advertisement

6. Other treatments

A notable therapeutic approach involves oncolytic viruses, which possess the ability to selectively destroy tumor cells while leaving healthy cells unharmed [217]. By causing the lysis of tumor cells, oncolytic viruses release TANs as well as tumor-associated antigens, thereby triggering a specific immune response. Rigvir, H101, and T-VEC are among the primary oncolytic viruses utilized in cancer therapy, with Rigvir being the pioneering oncolytic virus approved for clinical use [218].

Employing cytokines and non-specific immune boosters, which can be administered either before or concurrently with antigens to improve the immune response or modify its characteristics without inducing an antigenic response, represents an alternative approach for treating GC. Among these non-specific immune boosters, cytokines and lentinan are commonly employed in clinical settings. Lentinan, a β-1,3-D-glucan, is clinically employed as an immunomodulatory medication for tumor treatment [145].

Combination therapies are often used to achieve better treatment outcomes. For example, combining chemotherapy and trastuzumab in a clinical trial resulted in significantly longer OS rates. Gene therapy in combination with anti-cancer drugs presents a promising opportunity to enhance treatment outcomes [219]. An aptamer-siRNA chimera combined with 5-fluorouracil (5-FU) in a collagen membrane can specifically bind to GC cells, delivering 5-FU to the affected site while suppressing drug-resistant genes [220].

Advertisement

7. Conclusion and outlook

Gastric cancer (GC) is one of the prevalent cancers with poor prognosis. GC is responsible for a significant number of cancer-related deaths across the globe, particularly in its advanced stages (median survival of less than 1 year for metastatic patients). GC is known as a heterogeneous cancer, Furthermore, the biological differences between tumors in Eastern and Western countries have made finding a standard treatment that can include all types of GCs challenging. GC is a multifactorial disease caused by genetic and epigenetic factors; All these factors affect the biological regulatory processes of the cell such as proliferation, differentiation, and cell metabolism. In addition, tumor microenvironment (TEM) in GC differs from normal tissue regarding metabolism. These modifications include the shift from aerobic respiration to glycolysis which leads to lactate production and acidification of the TEM. The TEM contains a diverse range of cells, such as cancer cells, CAFs, pericytes, and other immune cells, and has irregular vasculature. Additionally, it is subject to various mutations. The conditions that were mentioned earlier have led to a lack of response to traditional forms of treatment like surgery, endoscopy, chemotherapy, and radiation therapy. The current treatment methods and the combined treatment of these methods, due to the resistance to the treatment, do not have the necessary effectiveness and are even associated with various side effects; Consequently, new treatment methods with biological approaches are felt more and more as a basic need to develop new treatment strategies. Studying tumor microenvironments and the interplay between cancer cells and immune cells has spurred the development of novel immunotherapy strategies, including the utilization of nanoparticles. A variety of immunotherapies including immune checkpoint inhibitors (CTLA-4 inhibitors،PD-1/PD-L1 inhibitors and Combinational therapy of Anti CTLA-4 and PD-1), cellular immunotherapy (NK cell transfer, TCR-T immunotherapy, and CAR-T cell therapies), cytokines and vaccine therapy have shown promising effects results against GC. NPs are utilized to actively target the tumor site. Moreover, morphological changes in the tumor TEM, such as variations in pH levels and distinct properties of its vessel wall, enable NPs to passively target cancer cells. Further, the usage of theranostic NPs, a combination of therapy and diagnosis, enables better monitoring of response during treatment. These capabilities allow NPs to effectively overcome limitations in GC therapy.

Despite the significant advancements made in the last decade, there are still many challenges to achieving a fully effective method with minimal side effects for the treatment of GC; including reducing the side effects caused by immunotherapy (colitis secondary to immunotherapy with diarrhea, abdominal pain, nausea), toxicity of nanomaterials, etc. In fact, it seems that we cannot eliminate conventional treatment methods, especially surgery and chemotherapy, anytime soon. In the near future, more efforts will be made to fully identify various biological regulatory factors such as ferroptosis, immunological GC, more effective factors on TEM, genomic data of different molecular subtypes, nanomaterials with appropriate biocompatibility and biodegradability. Additionally, scientists will be working to identify reliable predictive factors for early detection which will promise more effective treatment methods.

Advertisement

Author contributions

All authors contributed to the study conception, design, material preparation, data collection, and analysis. The first draft of the manuscript was written by and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

References

  1. 1. Januszewicz W, Turkot MH, Malfertheiner P, Regula J. A global perspective on gastric cancer screening: Which concepts are feasible, and when? Cancers. 2023;15(3):664
  2. 2. López MJ et al. Characteristics of gastric cancer around the world. Critical Reviews in Oncology/Hematology. 2023;181:103841
  3. 3. Ajani JA et al. Gastric cancer, version 2.2022, NCCN clinical practice guidelines in oncology. Journal of the National Comprehensive Cancer Network. 2022;20(2):167-192
  4. 4. Charalampakis N et al. Trimodality treatment in gastric and gastroesophageal junction cancers: Current approach and future perspectives. World Journal of Gastrointestinal Oncology. 2022;14(1):181
  5. 5. Jia Z et al. Positive H. Pylori status predicts better prognosis of non-cardiac gastric cancer patients: Results from cohort study and meta-analysis. BMC Cancer. 2022;22(1):155
  6. 6. Shin J, Park YS. Unusual or uncommon histology of gastric cancer. Journal of Gastric Cancer. 2024;24(1):69
  7. 7. Rawla P, Barsouk A. Epidemiology of gastric cancer: Global trends, risk factors and prevention. Gastroenterology Review/Przegląd Gastroenterologiczny. 2019;14(1):26-38
  8. 8. Mukaisho K, Nakayama T, Hagiwara T, Hattori T, Sugihara H. Two distinct etiologies of gastric cardia adenocarcinoma: Interactions among pH, Helicobacter pylori, and bile acids. Frontiers in Microbiology. 2015;6:412
  9. 9. Zhang Y et al. Healthy lifestyle counteracts the risk effect of genetic factors on incident gout: A large population-based longitudinal study. BMC Medicine. 2022;20(1):138
  10. 10. Sastre J, García-Saenz JA, Díaz-Rubio E. Chemotherapy for gastric cancer. World Journal of Gastroenterology: WJG. 2006;12(2):204
  11. 11. Högner A, Moehler M. Immunotherapy in gastric cancer. Current Oncology. 2022;29(3):1559-1574
  12. 12. Amjad MT, Chidharla A, Kasi A. Cancer chemotherapy. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2023. PMID: 33232037
  13. 13. Matsueda S, Graham DY. Immunotherapy in gastric cancer. World Journal of Gastroenterology: WJG. 2014;20(7):1657
  14. 14. Sah B et al. Post-operative complications of gastric cancer surgery: Female gender at high risk. European Journal of Cancer Care. 2009;18(2):202-208
  15. 15. Gao C, Liu F, Ye Q, Guo A. Cancer-associated fibroblasts affect tumor metabolism and immune microenvironment in gastric cancer and identification of its characteristic genes. Journal of Oncology. 2023;2023:1424589
  16. 16. Yan Y, Wang LF, Wang RF. Role of cancer-associated fibroblasts in invasion and metastasis of gastric cancer. World Journal of Gastroenterology. 2015;21(33):9717-9726
  17. 17. Sun H, Wang X, Wang X, Xu M, Sheng W. The role of cancer-associated fibroblasts in tumorigenesis of gastric cancer. Cell Death & Disease. 2022;13(10):874
  18. 18. Cirri P, Chiarugi P. Cancer associated fibroblasts: The dark side of the coin. American Journal of Cancer Research. 2011;1(4):482-497
  19. 19. Hanahan D, Coussens LM. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309-322
  20. 20. 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):135-147
  21. 21. Yamauchi M, Gibbons DL, Zong C, Fradette JJ, Bota-Rabassedas N, Kurie JM. Fibroblast heterogeneity and its impact on extracellular matrix and immune landscape remodeling in cancer. Matrix Biology. 2020;91-92:8-18
  22. 22. Mao X et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Molecular Cancer. 2021;20(1):131
  23. 23. Kumar V et al. Single-cell atlas of lineage states, tumor microenvironment, and subtype-specific expression programs in gastric cancer. Cancer Discovery. 2022;12(3):670-691
  24. 24. Li X et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics. 2022;12(2):620-638
  25. 25. Zhao Z, Mak TK, Shi Y, Li K, Huo M, Zhang C. Integrative analysis of cancer-associated fibroblast signature in gastric cancer. Heliyon. 2023;9(9):e19217
  26. 26. Rogers S et al. Cancer-associated fibroblasts influence Wnt/PCP signaling in gastric cancer cells by cytoneme-based dissemination of ROR2. Proceedings of the National Academy of Sciences of the United States of America. 2023;120(39):e2217612120
  27. 27. Kharaishvili G, Simkova D, Bouchalova K, Gachechiladze M, Narsia N, Bouchal J. The role of cancer-associated fibroblasts, solid stress and other microenvironmental factors in tumor progression and therapy resistance. Cancer Cell International. 2014;14:41
  28. 28. Sammarco G et al. Mast cells, angiogenesis and Lymphangiogenesis in human gastric cancer. International Journal of Molecular Sciences. 2019;20(9)
  29. 29. Xu X et al. Immunology and immunotherapy in gastric cancer. Clinical and Experimental Medicine. 2023;23(7):3189-3204
  30. 30. Zhao Y, Bai Y, Shen M, Li Y. Therapeutic strategies for gastric cancer targeting immune cells: Future directions. Frontiers in Immunology. 2022;13:992762
  31. 31. Yang J, Liu X, Cheng Y, Zhang J, Ji F, Ling Z. Roles of plasmacytoid dendritic cells in gastric cancer. Frontiers in Oncology. 2022;12:818314
  32. 32. Chang WJ, Du Y, Zhao X, Ma LY, Cao GW. Inflammation-related factors predicting prognosis of gastric cancer. World Journal of Gastroenterology. 2014;20(16):4586-4596
  33. 33. Wolf NK, Kissiov DU, Raulet DH. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nature Reviews. Immunology. 2023;23(2):90-105
  34. 34. Wu SY, Fu T, Jiang YZ, Shao ZM. Natural killer cells in cancer biology and therapy. Molecular Cancer. 2020;19(1):120
  35. 35. Fuertes MB, Domaica CI, Zwirner NW. Leveraging NKG2D ligands in Immuno-oncology. Frontiers in Immunology. 2021;12:713158
  36. 36. Devillier R, Chrétien AS, Pagliardini T, Salem N, Blaise D, Olive D. Mechanisms of NK cell dysfunction in the tumor microenvironment and current clinical approaches to harness NK cell potential for immunotherapy. Journal of Leukocyte Biology. 2021;109(6):1071-1088
  37. 37. Hu Z, Xu X, Wei H. The adverse impact of tumor microenvironment on NK-cell. Frontiers in Immunology. 2021;12:633361
  38. 38. van Vliet AA, Georgoudaki AM, Raimo M, de Gruijl TD, Spanholtz J. Adoptive NK cell therapy: A promising treatment Prospect for metastatic melanoma. Cancers (Basel). 2021;13(18)
  39. 39. Engström A, Erlandsson A, Delbro D, Wijkander J. Conditioned media from macrophages of M1, but not M2 phenotype, inhibit the proliferation of the colon cancer cell lines HT-29 and CACO-2. International Journal of Oncology. 2014;44(2):385-392
  40. 40. Arango Duque G, Descoteaux A. Macrophage cytokines: Involvement in immunity and infectious diseases. Frontiers in Immunology. 2014;5:491
  41. 41. Kurahara H et al. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. The Journal of Surgical Research. 2011;167(2):e211-e219
  42. 42. Li W et al. Gastric cancer-derived mesenchymal stromal cells trigger M2 macrophage polarization that promotes metastasis and EMT in gastric cancer. Cell Death & Disease. 2019;10(12):918
  43. 43. Kaparakis M et al. Macrophages are mediators of gastritis in acute Helicobacter pylori infection in C57BL/6 mice. Infection and Immunity. 2008;76(5):2235-2239
  44. 44. Sathe A et al. Single-cell genomic characterization reveals the cellular reprogramming of the gastric tumor microenvironment. Clinical Cancer Research. 2020;26(11):2640-2653
  45. 45. Eissmann MF et al. IL-33-mediated mast cell activation promotes gastric cancer through macrophage mobilization. Nature Communications. 2019;10(1):2735
  46. 46. Lin C et al. Tumour-associated macrophages-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut. 2019;68(10):1764-1773
  47. 47. Zhou Z et al. A C-X-C chemokine receptor type 2-dominated cross-talk between tumor cells and macrophages drives gastric cancer metastasis. Clinical Cancer Research. 2019;25(11):3317-3328
  48. 48. Li S et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. Journal of Experimental & Clinical Cancer Research. 2019;38(1):6
  49. 49. Wang TT et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut. 2017;66(11):1900-1911
  50. 50. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nature Reviews. Cancer. 2016;16(7):431-446
  51. 51. Mao Z et al. CXCL5 promotes gastric cancer metastasis by inducing epithelial-mesenchymal transition and activating neutrophils. Oncogene. 2020;9(7):63
  52. 52. Zhang W et al. Interaction with neutrophils promotes gastric cancer cell migration and invasion by inducing epithelial-mesenchymal transition. Oncology Reports. 2017;38(5):2959-2966
  53. 53. Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-derived suppressor cells as a therapeutic target for cancer. Cells. 2020;9(3)
  54. 54. Bayik D, Lee J, Lathia JD. The role of myeloid-derived suppressor cells in tumor growth and metastasis. Experientia Supplementum. 2022;113:189-217
  55. 55. Wang L, Chang EW, Wong SC, Ong SM, Chong DQ, Ling KL. Increased myeloid-derived suppressor cells in gastric cancer correlate with cancer stage and plasma S100A8/A9 proinflammatory proteins. Journal of Immunology. 2013;190(2):794-804
  56. 56. Cui C, Lan P, Fu L. The role of myeloid-derived suppressor cells in gastrointestinal cancer. Cancer Communications (London). 2021;41(6):442-471
  57. 57. Chen X et al. Accumulation of T-helper 22 cells, interleukin-22 and myeloid-derived suppressor cells promotes gastric cancer progression in elderly patients. Oncology Letters. 2018;16(1):253-261
  58. 58. Moaaz M, Lotfy H, Elsherbini B, Motawea MA, Fadali G. TGF-β enhances the anti-inflammatory effect of tumor-infiltrating CD33+11b+HLA-DR myeloid-derived suppressor cells in gastric cancer: A possible relation to MicroRNA-494. Asian Pacific Journal of Cancer Prevention. 2020;21(11):3393-3403
  59. 59. Wang PF et al. Prognostic role of pretreatment circulating MDSCs in patients with solid malignancies: A meta-analysis of 40 studies. Oncoimmunology. 2018;7(10):e1494113
  60. 60. Shen P, Fillatreau S. Antibody-independent functions of B cells: A focus on cytokines. Nature Reviews. Immunology. 2015;15(7):441-451
  61. 61. Sarvaria A, Madrigal JA, Saudemont A. B cell regulation in cancer and anti-tumor immunity. Cellular & Molecular Immunology. 2017;14(8):662-674
  62. 62. van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: Insights from single-cell analysis. Nature Reviews. Cancer. 2020;20(4):218-232
  63. 63. Basu A et al. Differentiation and regulation of T(H) cells: A balancing act for cancer immunotherapy. Frontiers in Immunology. 2021;12:669474
  64. 64. Kang BW, Kim JG, Lee IH, Bae HI, Seo AN. Clinical significance of tumor-infiltrating lymphocytes for gastric cancer in the era of immunology. World Journal of Gastrointestinal Oncology. 2017;9(7):293
  65. 65. Nagase H et al. ICOS(+) Foxp3(+) TILs in gastric cancer are prognostic markers and effector regulatory T cells associated with Helicobacter pylori. International Journal of Cancer. 2017;140(3):686-695
  66. 66. Miyara M et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30(6):899-911
  67. 67. Salama P et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. Journal of Clinical Oncology. 2009;27(2):186-192
  68. 68. Okita Y et al. Role of tumor-infiltrating CD11b+ antigen-presenting cells in the progression of gastric cancer. Journal of Surgical Research. 2014;186(1):192-200
  69. 69. Takeuchi H, Maehara Y, Tokunaga E, Koga T, Kakeji Y, Sugimachi K. Prognostic significance of natural killer cell activity in patients with gastric carcinoma: A multivariate analysis. The American Journal of Gastroenterology. 2001;96(2):574-578
  70. 70. Lee S-C et al. Phase I trial of expanded, activated autologous NK-cell infusions with trastuzumab in patients with HER2-positive cancers. Clinical Cancer Research. 2020;26(17):4494-4502
  71. 71. Ishigami S et al. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer. 2000;88(3):577-583
  72. 72. Xie M-Z, Ding K, Tang Y-P, Hu B-L, Li K-Z, Li J-L, et al. Percentage of natural killer (NK) cells in peripheral blood is associated with prognosis in patients with gastric cancer: A retrospective study from a single center. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research. 2021;27:e927464-e927461
  73. 73. Guo Z, Zhou C, Zhou L, Wang Z, Zhu X, Mu X. Overexpression of DAPK1-mediated inhibition of IKKβ/CSN5/PD-L1 axis enhances natural killer cell killing ability and inhibits tumor immune evasion in gastric cancer. Cellular Immunology. 2022;372:104469
  74. 74. Pernot S et al. Infiltrating and peripheral immune cell analysis in advanced gastric cancer according to the Lauren classification and its prognostic significance. Gastric Cancer. 2020;23:73-81
  75. 75. Fabian KP et al. PD-L1 targeting high-affinity NK (t-haNK) cells induce direct antitumor effects and target suppressive MDSC populations. Journal for Immunotherapy of Cancer. 2020;8(1)
  76. 76. Yu S et al. Activated HIF1α of tumor cells promotes chemoresistance development via recruiting GDF15-producing tumor-associated macrophages in gastric cancer. Cancer Immunology, Immunotherapy. 2020;69:1973-1987
  77. 77. Lin C et al. Tumour-associated macrophages-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut. 2019. DOI: 10.1136/gutjnl-2018-316324
  78. 78. Peng L-S et al. Tumor-associated monocytes/macrophages impair NK-cell function via TGFβ1 in human gastric cancer. Cancer Immunology Research. 2017;5(3):248-256
  79. 79. Wang H-C et al. Tumor-associated macrophages promote epigenetic silencing of gelsolin through DNA methyltransferase 1 in gastric cancer cells. Cancer Immunology Research. 2017;5(10):885-897
  80. 80. Liu X et al. Poor clinical outcomes of intratumoral dendritic cell–specific intercellular adhesion molecule 3–grabbing non-integrin–positive macrophages associated with immune evasion in gastric cancer. European Journal of Cancer. 2020;128:27-37
  81. 81. Jeremiasen M et al. Tumor-associated CD68+, CD163+, and MARCO+ macrophages as prognostic biomarkers in patients with treatment-naïve gastroesophageal adenocarcinoma. Frontiers in Oncology. 2020;10:534761
  82. 82. Zhang H et al. Poor clinical outcomes and immunoevasive contexture in intratumoral IL-10-producing macrophages enriched gastric cancer patients. Annals of Surgery. 2022;275(4):e626-e635
  83. 83. Zhang X, Shi H, Yuan X, Jiang P, Qian H, Xu W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Molecular Cancer. 2018;17(1):146
  84. 84. Li TJ et al. Interleukin-17-producing neutrophils link inflammatory stimuli to disease progression by promoting angiogenesis in gastric cancer. Clinical Cancer Research. 2017;23(6):1575-1585
  85. 85. Zhou X et al. PMN-MDSCs accumulation induced by CXCL1 promotes CD8+ T cells exhaustion in gastric cancer. Cancer Letters. 2022;532:215598
  86. 86. Thompson ED et al. Patterns of PD-L1 expression and CD8 T cell infiltration in gastric adenocarcinomas and associated immune stroma. Gut. 2016. gutjnl-2015-310839. DOI: 10.1016/j.canlet.2022.215598
  87. 87. He X et al. Impact of intratumoural CD73 expression on prognosis and therapeutic response in patients with gastric cancer. European Journal of Cancer. 2021;157:114-123
  88. 88. Wang J et al. Intratumoral CXCR5+ CD8+ T associates with favorable clinical outcomes and immunogenic contexture in gastric cancer. Nature Communications. 2021;12(1):3080
  89. 89. Jin K et al. Poor clinical outcomes and immunoevasive contexture in CXCL13+ CD8+ T cells enriched gastric cancer patients. Oncoimmunology. 2021;10(1):1915560
  90. 90. Nagase H et al. ICOS+ F oxp3+ TIL s in gastric cancer are prognostic markers and effector regulatory T cells associated with Helicobacter pylori. International Journal of Cancer. 2017;140(3):686-695
  91. 91. Fei Y et al. Intratumoral Foxp3+ RORγt+ T cell infiltration determines poor prognosis and immunoevasive contexture in gastric cancer patients. Cancer Immunology, Immunotherapy. 2022;71(1):1-11
  92. 92. Wang J et al. Tumor-infiltrating γδT cells predict prognosis and adjuvant chemotherapeutic benefit in patients with gastric cancer. Oncoimmunology. 2017;6(11):e1353858
  93. 93. Lin R et al. Fatty acid oxidation controls CD8+ tissue-resident memory T-cell survival in gastric adenocarcinoma. Cancer Immunology Research. 2020;8(4):479-492
  94. 94. Kwak Y et al. Differential prognostic impact of CD8+ T cells based on human leucocyte antigen I and PD-L1 expression in microsatellite-unstable gastric cancer. British Journal of Cancer. 2020;122(9):1399-1408
  95. 95. Chen K et al. TIM3+ cells in gastric cancer: Clinical correlates and association with immune context. British Journal of Cancer. 2022;126(1):100-108
  96. 96. Qu Y et al. The effects of TNF-α/TNFR2 in regulatory T cells on the microenvironment and progression of gastric cancer. International Journal of Cancer. 2022;150(8):1373-1391
  97. 97. Zhu F et al. Ring finger protein 31–mediated atypical ubiquitination stabilizes forkhead box P3 and thereby stimulates regulatory T-cell function. Journal of Biological Chemistry. 2018;293(52):20099-20111
  98. 98. Mao F-Y et al. Increased tumor-infiltrating CD45RA−CCR7−regulatory T-cell subset with immunosuppressive properties foster gastric cancer progress. Cell Death & Disease. 2017;8(8):e3002
  99. 99. Kindlund B et al. CD4+ regulatory T cells in gastric cancer mucosa are proliferating and express high levels of IL-10 but little TGF-β. Gastric Cancer. 2017;20:116-125
  100. 100. Liu X et al. Regulatory T cells and M2 macrophages present diverse prognostic value in gastric cancer patients with different clinicopathologic characteristics and chemotherapy strategies. Journal of Translational Medicine. 2019;17(1):1-11
  101. 101. Ni Z et al. Tumor-infiltrating B cell is associated with the control of progression of gastric cancer. Immunologic Research. 2021;69:43-52
  102. 102. Ji L et al. Blockade of β-catenin-induced CCL28 suppresses gastric cancer progression via inhibition of Treg cell infiltration. Cancer Research. 2020;80(10):2004-2016
  103. 103. Fang H et al. Intratumoral interleukin-9 delineates a distinct immunogenic class of gastric cancer patients with better prognosis and adjuvant chemotherapeutic response. Oncoimmunology. 2020;9(1):1856468
  104. 104. Sun L et al. Human gastric cancer mesenchymal stem cell-derived IL15 contributes to tumor cell epithelial-mesenchymal transition via upregulation Tregs ratio and PD-1 expression in CD4(+)T cell. Stem Cells and Development. 2018;27(17):1203-1214
  105. 105. Wang JT et al. Intratumoral IL17-producing cells infiltration correlate with antitumor immune contexture and improved response to adjuvant chemotherapy in gastric cancer. Annals of Oncology. 2019;30(2):266-273
  106. 106. Bie Q et al. IL-17B/IL-17RB signaling cascade contributes to self-renewal and tumorigenesis of cancer stem cells by regulating Beclin-1 ubiquitination. Oncogene. 2021;40(12):2200-2216
  107. 107. Wu P et al. Adaptive mechanisms of tumor therapy resistance driven by tumor microenvironment. Frontiers in Cell and Developmental Biology. 2021;9:641469
  108. 108. Subhash VV, Yeo MS, Tan WL, Yong WP. Strategies and advancements in harnessing the immune system for gastric cancer immunotherapy. Journal of Immunology Research, vol. 2015;2015
  109. 109. Pettersen EO et al. Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: The METOXIA consortium. Journal of Enzyme Inhibition and Medicinal Chemistry. 2015;30(5):689-721
  110. 110. Al Tameemi W, Dale TP, Al-Jumaily RMK, Forsyth NR. Hypoxia-modified cancer cell metabolism. Frontiers in Cell and Developmental Biology. 2019;7:4
  111. 111. Das V, Štěpánková J, Hajdúch M, Miller JH. Role of tumor hypoxia in acquisition of resistance to microtubule-stabilizing drugs. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2015;1855(2):172-182
  112. 112. Li M, Li G, Yang X, Yin W, Lv G, Wang S. HIF in gastric cancer: Regulation and therapeutic target. Molecules. 2022;27(15):4893
  113. 113. Griffiths EA, Pritchard S, Welch I, Price PM, West C. Is the hypoxia-inducible factor pathway important in gastric cancer? European Journal of Cancer. 2005;41(18):2792-2805
  114. 114. Xuan Y, Wang YN. Hypoxia/IL-1α axis promotes gastric cancer progression and drug resistance. Journal of Digestive Diseases. 2017;18(9):511-520
  115. 115. Sun X et al. Hypoxia-mediated cancer stem cell resistance and targeted therapy. Biomedicine & Pharmacotherapy. 2020;130:110623
  116. 116. Kang BW, Chau I. Current status and future potential of predictive biomarkers for immune checkpoint inhibitors in gastric cancer. ESMO Open. 2020;5(4):e000791
  117. 117. Liu Y et al. Signaling pathways of oxidative stress response: The potential therapeutic targets in gastric cancer. Frontiers in Immunology. 2023;14:1139589
  118. 118. Gu H et al. Reactive oxygen species-mediated tumor microenvironment transformation: The mechanism of radioresistant gastric cancer. Oxidative Medicine and Cellular Longevity. 2018;2018
  119. 119. Allegra A, Murdaca G, Mirabile G, Gangemi S. Redox Signaling modulates activity of immune checkpoint inhibitors in cancer patients. Biomedicine. 2023;11(5):1325
  120. 120. Aboelella NS, Brandle C, Kim T, Ding Z-C, Zhou G. Oxidative stress in the tumor microenvironment and its relevance to cancer immunotherapy. Cancers. 2021;13(5):986
  121. 121. Leone A, Roca MS, Ciardiello C, Costantini S, Budillon A. Oxidative stress gene expression profile correlates with cancer patient poor prognosis: Identification of crucial pathways might select novel therapeutic approaches. Oxidative Medicine and Cellular Longevity. 2017;2017
  122. 122. Justus CR, Sanderlin EJ, Yang LV. Molecular connections between cancer cell metabolism and the tumor microenvironment. International Journal of Molecular Sciences. 2015;16(5):11055-11086
  123. 123. Chiche J, Brahimi-Horn MC, Pouysségur J. Tumour hypoxia induces a metabolic shift causing acidosis: A common feature in cancer. Journal of Cellular and Molecular Medicine. 2010;14(4):771-794
  124. 124. Wu H et al. T-cells produce acidic niches in lymph nodes to suppress their own effector functions. Nature Communications. 2020;11(1):4113
  125. 125. Colombani T et al. Oxygen-generating cryogels restore T cell mediated cytotoxicity in hypoxic tumors. Advanced Functional Materials. 2021;31(37):2102234
  126. 126. Ramello MC, Haura EB, Abate-Daga D. CAR-T cells and combination therapies: what’s next in the immunotherapy revolution? Pharmacological Research. 2018;129:194-203
  127. 127. Bai J, Gao Z, Li X, Dong L, Han W, Nie J. Regulation of PD-1/PD-L1 pathway and resistance to PD-1/PD-L1 blockade. Oncotarget. 2017;8(66):110693
  128. 128. Wu Y, Chen W, Xu ZP, Gu W. PD-L1 distribution and perspective for cancer immunotherapy—Blockade, knockdown, or inhibition. Frontiers in Immunology. 2019;10:2022
  129. 129. Petryszyn P, Chapelle N, Matysiak-Budnik T. Gastric cancer: Where are we heading? Digestive Diseases. 2020;38(4):280-285
  130. 130. Arn CR, Halla KJ, Gill S. Tisotumab Vedotin safety and tolerability in clinical practice: Managing adverse events. Journal of the Advanced Practitioner in Oncology. 2023;14(2):139
  131. 131. Galata C, Ronellenfitsch U, Blank S, Reißfelder C, Hardt J. Postoperative morbidity and failure to rescue in surgery for gastric cancer: A single center retrospective cohort study of 1107 patients from 1972 to 2014. Cancers. 2020;12(7):1953
  132. 132. Das M. Neoadjuvant chemotherapy: Survival benefit in gastric cancer. The Lancet Oncology. 2017;18(6):e307
  133. 133. Maisey N, Norman A, Prior Y, Cunningham D. Chemotherapy for primary gastric lymphoma: Does in-patient observation prevent complications? Clinical Oncology. 2004;16(1):48-52
  134. 134. Natale JJ. Overview of the prevention and management of CINV. The American Journal of Managed Care. 2018;24(18 Suppl.):S391-S397
  135. 135. Andreyev HJN, Davidson SE, Gillespie C, Allum WH, Swarbrick E. Practice guidance on the management of acute and chronic gastrointestinal problems arising as a result of treatment for cancer. Gut. 2011. gutjnl-2011-300563. DOI: 10.1016/S1470-2045(17)30321-2
  136. 136. Ng SP, Leong T. Role of radiation therapy in gastric cancer. Annals of Surgical Oncology. 2021;28(8):4151-4157
  137. 137. Kennedy GD, Heise CP. Radiation colitis and proctitis. Clinics in Colon and Rectal Surgery. 2007;20(01):064-072
  138. 138. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nature Reviews Drug Discovery. 2019;18(3):175-196
  139. 139. Zhao Q et al. Immunotherapy for gastric cancer: Dilemmas and prospect. Briefings in Functional Genomics. 2019;18(2):107-112
  140. 140. Grierson P, Lim K-H, Amin M. Immunotherapy in gastrointestinal cancers. Journal of Gastrointestinal Oncology. 2017;8(3):474
  141. 141. De Velasco G et al. Comprehensive meta-analysis of key immune-related adverse events from CTLA-4 and PD-1/PD-L1 inhibitors in cancer patients. Cancer Immunology Research. 2017;5(4):312-318
  142. 142. Larkin J et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. New England Journal of Medicine. 2015;373(1):23-34
  143. 143. Marthey L et al. Cancer immunotherapy with anti-CTLA-4 monoclonal antibodies induces an inflammatory bowel disease. Journal of Crohn's and Colitis. 2016;10(4):395-401
  144. 144. Akkanapally V, Bai X-F, Basu S. Therapeutic immunomodulation in gastric cancer. Cancers. 2024;16(3):560
  145. 145. Li K, Zhang A, Li X, Zhang H, Zhao L. Advances in clinical immunotherapy for gastric cancer. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2021;1876(2):188615
  146. 146. Chen L, Han X. Anti–PD-1/PD-L1 therapy of human cancer: Past, present, and future. The Journal of Clinical Investigation. 2015;125(9):3384-3391
  147. 147. Park J-J et al. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood, The Journal of the American Society of Hematology. 2010;116(8):1291-1298
  148. 148. Takei S, Kawazoe A, Shitara K. The new era of immunotherapy in gastric cancer. Cancers. 2022;14(4):1054
  149. 149. Miljanic M, Capasso A, Triplett TA, Eckhardt SG, Aung KL. Immune checkpoint blockade in gastrointestinal cancers: The current status and emerging paradigms. Journal of Immunotherapy and Precision Oncology. 2020;3(1):3-15
  150. 150. Shimozaki K, Nakayama I, Hirota T, Yamaguchi K. Current strategy to treat immunogenic gastrointestinal cancers: Perspectives for a new era. Cells. 2023;12(7):1049
  151. 151. Sznol M, Melero I. Revisiting anti-CTLA-4 antibodies in combination with PD-1 blockade for cancer immunotherapy. Annals of Oncology. 2021;32(3):295-297
  152. 152. Janjigian YY et al. CheckMate-032: Phase I/II, open-label study of safety and activity of nivolumab (nivo) alone or with ipilimumab (ipi) in advanced and metastatic (A/M) gastric cancer (GC). Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 2016;1876(2):188615
  153. 153. Kelly RJ et al. Safety and efficacy of durvalumab and tremelimumab alone or in combination in patients with advanced gastric and gastroesophageal junction adenocarcinoma. Clinical Cancer Research. 2020;26(4):846-854
  154. 154. Shitara K et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial. The Lancet. 2018;392(10142):123-133
  155. 155. Moehler M et al. Phase III trial of avelumab maintenance after first-line induction chemotherapy versus continuation of chemotherapy in patients with gastric cancers: Results from JAVELIN gastric 100. Journal of Clinical Oncology. 2021;39(9):966
  156. 156. Li K, Zhang A, Li X, Zhang H, Zhao L. Advances in clinical immunotherapy for gastric cancer. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 2021;1876(2):188615
  157. 157. Yu Y et al. Changes in expression of multiple checkpoint molecules and infiltration of tumor immune cells after neoadjuvant chemotherapy in gastric cancer. Journal of Cancer. 2019;10(12):2754
  158. 158. Muro K et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): A multicentre, open-label, phase 1b trial. The Lancet Oncology. 2016;17(6):717-726
  159. 159. Kang Y-K et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): A randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2017;390(10111):2461-2471
  160. 160. Wang B, Qin L, Ren M, Sun H. Effects of combination of anti-CTLA-4 and anti-PD-1 on gastric cancer cells proliferation, apoptosis and metastasis. Cellular Physiology and Biochemistry. 2018;49(1):260-270
  161. 161. Du Y, Wei Y. Therapeutic potential of natural killer cells in gastric cancer. Frontiers in Immunology. 2019;9:3095
  162. 162. Sakamoto N et al. Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. Journal of Translational Medicine. 2015;13:1-13
  163. 163. Alcantara M, Du Rusquec P, Romano E. Current clinical evidence and potential solutions to increase benefit of CAR T-cell therapy for patients with solid tumors. Oncoimmunology. 2020;9(1):1777064
  164. 164. Bębnowska D et al. CAR-T cell therapy—An overview of targets in gastric cancer. Journal of Clinical Medicine. 2020;9(6):1894
  165. 165. Zhan X et al. Phase I trial of Claudin 18.2-specific chimeric antigen receptor T cells for advanced gastric and pancreatic adenocarcinoma. Journal of Translational Medicine. 2019;13(1):277
  166. 166. Jiang H et al. Claudin18. 2-specific chimeric antigen receptor engineered T cells for the treatment of gastric cancer. JNCI: Journal of the National Cancer Institute. 2019;111(4):409-418
  167. 167. Tao K et al. Development of NKG2D-based chimeric antigen receptor-T cells for gastric cancer treatment. Cancer Chemotherapy and Pharmacology. 2018;82:815-827
  168. 168. Grillo F, Fassan M, Sarocchi F, Fiocca R, Mastracci L. HER2 heterogeneity in gastric/gastroesophageal cancers: From benchside to practice. World Journal of Gastroenterology. 2016;22(26):5879
  169. 169. Song Y et al. Effective and persistent antitumor activity of HER2-directed CAR-T cells against gastric cancer cells in vitro and xenotransplanted tumors in vivo. Protein & Cell. 2018;9(10):867-878
  170. 170. Kim M et al. Folate receptor 1 (FOLR1) targeted chimeric antigen receptor (CAR) T cells for the treatment of gastric cancer. PLoS One. 2018;13(6):e0198347
  171. 171. Maude SL et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. New England Journal of Medicine. 2018;378(5):439-448
  172. 172. Lv J et al. Mesothelin is a target of chimeric antigen receptor T cells for treating gastric cancer. Journal of Hematology & Oncology. 2019;12(1):1-14
  173. 173. Jahn L et al. TCR-based therapy for multiple myeloma and other B-cell malignancies targeting intracellular transcription factor BOB1. Blood. 2017;129(10):1284-1295
  174. 174. Robbins PF et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: Long-term follow-up and correlates with response. Clinical Cancer Research. 2015;21(5):1019-1027
  175. 175. Xia Y et al. Treatment of metastatic non-small cell lung cancer with NY-ESO-1 specific TCR engineered-T cells in a phase I clinical trial: A case report. Oncology Letters. 2018;16(6):6998-7007
  176. 176. Ichiki Y et al. Development of adoptive immunotherapy with KK-LC-1-specific TCR-transduced γδT cells against lung cancer cells. Cancer Science. 2020;111(11):4021-4030
  177. 177. Shida A et al. Cancer/testis antigen, Kita-Kyushu lung cancer antigen-1 and ABCD stratification for diagnosing gastric cancers. World Journal of Gastroenterology. 2020;26(4):424-432
  178. 178. Marcinkowski B et al. Cancer targeting by TCR gene-engineered T cells directed against Kita-Kyushu lung cancer Antigen-1. Journal for Immunotherapy of Cancer (MDPI). 2019;7(1):229
  179. 179. Fujiwara S et al. NY-ESO-1 antibody as a novel tumour marker of gastric cancer. British Journal of Cancer. 2013;108(5):1119-1125
  180. 180. Watson HA et al. L-selectin enhanced T cells improve the efficacy of cancer immunotherapy. Frontiers in Immunology. 2019;10:1321
  181. 181. Zhang K et al. Phase II trial of adjuvant immunotherapy with autologous tumor-derived Gp96 vaccination in patients with gastric cancer. Journal of Cancer. 2017;8(10):1826-1832
  182. 182. Subklewe M, von Bergwelt-Baildon M, Humpe A. Chimeric antigen receptor T cells: A race to revolutionize cancer therapy. Transfusion Medicine and Hemotherapy. 2019;46(1):15-24
  183. 183. Ishikawa H et al. Phase I clinical trial of vaccination with LY6K-derived peptide in patients with advanced gastric cancer. Gastric Cancer. 2014;17(1):173-180
  184. 184. Hani U et al. Novel drug delivery systems as an emerging platform for stomach cancer therapy. Pharmaceutics. 2022;14(8):1576
  185. 185. Zielińska A et al. Polymeric nanoparticles: Production, characterization, toxicology and ecotoxicology. Molecules. 2020;25(16)
  186. 186. Hosseini S, Mohammadnejad J, Salamat S, Zadeh ZB, Tanhaei M, Ramakrishna S. Theranostic polymeric nanoparticles as a new approach in cancer therapy and diagnosis: A review. Materials Today Chemistry. 2023;29:101400
  187. 187. Suriya Prabha A, Dorothy R, Jancirani S, Rajendran S, Singh G, Senthil Kumaran S. In: Nanotoxicity S, Rajendran A, Mukherjee TA, Nguyen CG, Shukla RK, editors. Chapter 7 - Recent Advances in the Study of Toxicity of Polymer-Based Nanomaterials. Elsevier; 2020. pp. 143-165
  188. 188. Hong J, Feng Z. Synergic fabrication of combination therapy of irinotecan and 5-fluorouracil encapsulated polymeric nanoparticles for the treatment of gastric cancer therapy. Process Biochemistry. 2021;106:191-198
  189. 189. Narmani A et al. Biomedical applications of PLGA nanoparticles in nanomedicine: Advances in drug delivery systems and cancer therapy. Expert Opinion on Drug Delivery. no. just-accepted. 2023
  190. 190. Hosseini SM, Mohammadnejad J, Yousefnia H, Alirezapour B, Rezayan AH. Development of 177Lu-Cetuximab-PAMAM dendrimeric nanosystem: A novel theranostic radioimmunoconjugate. Journal of Cancer Research and Clinical Oncology. 2023;149(10):7779-7791
  191. 191. Pan L et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. Journal of Cancer Research and Clinical Oncology. 2017;143(6):991-1004
  192. 192. Fu M, Gu J, Jiang P, Qian H, Xu W, Zhang X. Exosomes in gastric cancer: Roles, mechanisms, and applications. Molecular Cancer. 2019;18:1-12
  193. 193. Das M, Huang L. Liposomal nanostructures for drug delivery in gastrointestinal cancers. Journal of Pharmacology and Experimental Therapeutics. 2019;370(3):647-656
  194. 194. Pattni BS, Chupin VV, Torchilin VP. New developments in liposomal drug delivery. Chemical Reviews. 2015;115(19):10938-10966
  195. 195. Sharma A, Goyal AK, Rath G. Recent advances in metal nanoparticles in cancer therapy. Journal of Drug Targeting. 2018;26(8):617-632
  196. 196. Salapa J, Bushman A, Lowe K, Irudayaraj J. Nano drug delivery systems in upper gastrointestinal cancer therapy. Nano Convergence. 2020;7(1):1-17
  197. 197. Hosseinkhah M et al. Cytotoxic potential of nickel oxide nanoparticles functionalized with glutamic acid and conjugated with thiosemicarbazide (NiO@ Glu/TSC) against human gastric cancer cells. Journal of Cluster Science. 2022;33(5):2045-2053
  198. 198. Jarestan M et al. Preparation, characterization, and anticancer efficacy of novel cobalt oxide nanoparticles conjugated with thiosemicarbazide. 3 Biotech. 2020;10:1-9
  199. 199. Shi H et al. Magnetic semiconductor Gd-doping CuS nanoparticles as Activatable Nanoprobes for bimodal imaging and targeted Photothermal therapy of gastric Tumors. Nano Letters. 2019;19(2):937-947
  200. 200. Hosseini SM, Mohammadnejad J, Najafi-Taher R, Zadeh ZB, Tanhaei M, Ramakrishna S. Multifunctional carbon-based nanoparticles: Theranostic applications in cancer therapy and diagnosis. ACS Applied Bio Materials. 2023;6(4):1323-1338
  201. 201. Deng L, Guo W, Li G, Hu Y, Zhang L-M. Hydrophobic IR780 loaded sericin nanomicelles for phototherapy with enhanced antitumor efficiency. International Journal of Pharmaceutics. 2019;566:549-556
  202. 202. Tsujimoto H et al. Theranostic photosensitive nanoparticles for lymph node metastasis of gastric cancer. Annals of Surgical Oncology. 2015;22(3):923-928
  203. 203. Zhang A et al. Carbon-gold hybrid nanoprobes for real-time imaging, photothermal/photodynamic and nanozyme oxidative therapy. Theranostics. 2019;9(12):3443
  204. 204. Shi H et al. Tumor-targeting CuS nanoparticles for multimodal imaging and guided photothermal therapy of lymph node metastasis. Acta Biomaterialia. 2018;72:256-265
  205. 205. Liu D et al. Target-specific delivery of oxaliplatin to HER2-positive gastric cancer cells in vivo using oxaliplatin-au-fe3o4-herceptin nanoparticles. Oncology Letters. 2018;15(5):8079-8087
  206. 206. Yang Z et al. Tumor-targeting W18O49 nanoparticles for dual-modality imaging and guided heat-shock-response-inhibited photothermal therapy in gastric cancer. Particle & Particle Systems Characterization. 2019;36(7):1900124
  207. 207. Yang Z et al. Defeating relapsed and refractory malignancies through a nano-enabled mitochondria-mediated respiratory inhibition and damage pathway. Biomaterials. 2020;229:119580
  208. 208. Yao H, Xu K, Zhou J, Zhou L, Wei S. A tumor microenvironment destroyer for efficient cancer suppression. ACS Biomaterials Science & Engineering. 2019;6(1):450-462
  209. 209. Chen J et al. CuS–NiS2 nanomaterials for MRI guided phototherapy of gastric carcinoma via triggering mitochondria-mediated apoptosis and MLKL/CAPG-mediated necroptosis. Nanotoxicology. 2020;14(6):774-787
  210. 210. Abtab SMT et al. Reticular chemistry in action: A hydrolytically stable MOF capturing twice its weight in adsorbed water. Chem. 2018;4(1):94-105
  211. 211. Zhou Y, Sun X, Zhou L, Zhang X. pH-sensitive and long-circulation nanoparticles for near-infrared fluorescence imaging-monitored and chemo-photothermal synergistic treatment against gastric cancer. Frontiers in Pharmacology. 2020;11:610883
  212. 212. Meng X et al. Photothermal/photodynamic therapy with immune-adjuvant liposomal complexes for effective gastric cancer therapy. Particle & Particle Systems Characterization. 2019;36(6):1900015
  213. 213. Shi T et al. Enhanced legumain-recognition and NIR controlled released of cisplatin-indocyanine nanosphere against gastric carcinoma. European Journal of Pharmacology. 2017;794:184-192
  214. 214. Li X, Ai S, Lu X, Liu S, Guan W. Nanotechnology-based strategies for gastric cancer imaging and treatment. RSC Advances. 2021;11(56):35392-35407
  215. 215. Arshad R et al. Novel perspectives towards RNA-based nano-theranostic approaches for cancer management. Nanomaterials. 2021;11(12):3330
  216. 216. Luo X, Peng X, Hou J, Wu S, Shen J, Wang L. Folic acid-functionalized polyethylenimine superparamagnetic iron oxide nanoparticles as theranostic agents for magnetic resonance imaging and PD-L1 siRNA delivery for gastric cancer. International Journal of Nanomedicine. 2017;11:5331-5343
  217. 217. Ahmadi A, Ghaleh HE, Dorostkar R, Farzanehpour M, Bolandian M. Oncolytic Coxsackievirus and the mechanisms of its effects on cancer: A narrative review. Current Cancer Therapy Reviews. 2021;17(3):173-178
  218. 218. Chen L, Zuo M, Zhou Q, Wang Y. Oncolytic virotherapy in cancer treatment: Challenges and optimization prospects. Frontiers in Immunology. 2023;14:1308890
  219. 219. Dai X, Tan C. Combination of microRNA therapeutics with small-molecule anticancer drugs: Mechanism of action and co-delivery nanocarriers. Advanced Drug Delivery Reviews. 2015;81:184-197
  220. 220. Chen W et al. Construction of Aptamer-siRNA chimera/PEI/5-FU/carbon nanotube/collagen membranes for the treatment of peritoneal dissemination of drug-resistant gastric cancer. Advanced Healthcare Materials. 2020;9(21):2001153

Written By

Pegah Mousavi, Ali Ahmadi, Shakila Behzadifar, Javad Mohammadnejad and Seyed Mohammad Hosseini

Submitted: 01 March 2024 Reviewed: 11 March 2024 Published: 01 July 2024