Home The tumor microenvironment: a key player in multidrug resistance in cancer
Article Open Access

The tumor microenvironment: a key player in multidrug resistance in cancer

  • Lingnan Meng , Ying Zheng , Hao Liu and Daiming Fan EMAIL logo
Published/Copyright: January 3, 2024

Abstract

Cancer is the second leading cause of death worldwide. Although multiple new cancer treatments have emerged in recent years, drug therapy, mainly comprising chemotherapy, targeted therapy, and immunotherapy, remains the most common approach. The multidrug resistance (MDR) of cancer cells to various treatments remains a challenge. Scientists have always focused on the acquired drug resistance mechanisms of tumor cells themselves. However, recent evidence shows that the tumor microenvironment (TME) plays a critical role in regulating tumor cell progression, metastasis, immune escape, and drug resistance. In the TME, interactions between cancer cells and non-malignant cells often modify the TME and facilitate drug resistance. Therefore, elucidating this complex interaction mechanism is essential for the development of effective treatments. This review focuses on the role of the TME in promoting chemoresistance in tumor cells through the following mechanisms: (i) inhibiting the immune clearance of tumor cells and facilitating immune escape responses; (ii) stimulating the release of soluble paracrine factors to enhance tumor survival and growth; (iii) promoting survival and altering drug delivery through metabolic reprogramming; (iv) obstructing drug absorption by inducing changes in stomatal cells and blood vessels surrounding the tumor; and (v) inducing the cancer stem cell phenotype. This review also addresses a clinical treatment strategy for targeting the TME, providing insights and a basis for reversing multidrug resistance.

Introduction

According to the latest data from the World Health Organization, the incidence of cancer cases is projected to increase by approximately 60 % in the next 20 years [1, 2]. Approximately one in six people die from cancer each year, making it the second leading cause of death worldwide [3]. As a result, cancer treatment remains a significant concern. Anti-cancer treatments include surgery, radiotherapy, chemotherapy, and immunotherapy [4]. In recent years, new treatments have emerged, and more drugs have been incorporated into clinical practice. However, several factors limit the effectiveness of cancer treatment, and numerous challenges still exist.

The use of anti-cancer drugs is crucial in clinical practice, but resistance often develops, particularly when palliative care is given to patients with advanced or metastatic solid cancers, ultimately leading to disease progression and death [5], [6], [7]. Cancer cells can develop resistance to drugs that interact with tumor cells during long-term drug therapy, and multidrug resistance (MDR) refers to cross-resistance to drugs of various structures and mechanisms of action to which they have not been exposed [8]. The effectiveness of clinical anticancer therapy is limited by the challenging issue of MDR. Previous studies on the mechanisms of MDR have primarily examined intrinsic cellular heterogeneity, increased drug efflux, and genetic or phenotypic changes. However, more recent research has highlighted the crucial role of the tumor microenvironment (TME) in determining therapeutic resistance in tumors [9, 10].

The TME is a complex and integrated system that primarily comprises tumor cells, their surrounding immune and inflammatory cells in their vicinity, tumor-associated fibroblasts (TAFs) and adjacent mesenchymal tissue, microvasculature, and various cytokines and chemokines [11, 12]. Tumor cells can be thought of as the seeds, and TMEs as the soil. They interact and integrate to form a metabolic microenvironment characterized by immunosuppression and nutritional deficiencies [13]. This unfavorable metabolic environment not only facilitates the growth, spread, and other harmful changes in tumor cells, but also triggers a local inflammatory response within the tumor. This response hinders the destruction of the tumor by immune cells and ultimately leads to the development of drug resistance [14], [15], [16], [17], [18], [19], [20] (Figure 1).

Figure 1: 
Effects of the TME on drug resistance. The TME is a micro-ecosystem comprising extracellular matrix (ECM), TAFs, soluble factors, endothelial cells, tumor-associated macrophages (TAMs), EVs, and adaptive and innate immune cells, including dendritic cells (DCs), B cells, T cells, and cytotoxic and regulatory T cells. The figure was set up using BioRender.com.
Figure 1:

Effects of the TME on drug resistance. The TME is a micro-ecosystem comprising extracellular matrix (ECM), TAFs, soluble factors, endothelial cells, tumor-associated macrophages (TAMs), EVs, and adaptive and innate immune cells, including dendritic cells (DCs), B cells, T cells, and cytotoxic and regulatory T cells. The figure was set up using BioRender.com.

There is a growing understanding of the critical function of TME in tumor formation and treatment resistance. Targeting the TME has recently emerged as a novel approach to treat MDR cancers [21]. Targeting specific components of TME to modulate the immune, and trophic metabolism of TME can reprogram the immune response and overcome drug resistance. This has the potential to provide maximum therapeutic benefit for cancer patients [22]. Therefore, it is necessary to have a better understanding of the factors in TME that promote MDR to achieve targeted therapy for MDR cancers by targeting TME. This paper discusses and analyses TME and its effects on MDR and its potential mechanisms. It also describes clinical trials conducted for TME.

Role of the TME in tumor drug resistance

Possible interactions between tumor cells and components of TME [23]. Several components in the TME can contribute to MDR in tumor cells through various mechanisms: (i) preventing immune clearance of tumor cells and promoting immunological escape [24], [25], [26], [27]; (ii) secreting soluble factors in a paracrine manner to promote survival and tumor growth [27, 28]; (iii) increasing survival and altering drug delivery through metabolic reprogramming [29], [30], [31]; (iv) altering the stomatal cells and blood arteries surrounding the tumor to prevent medication absorption; and (v) causing the phenotype of cancer stem cells (CSCs) [32], [33], [34].

The role of immunosuppressive components in TME

Numerous immune cells infiltrate the TME, including cytotoxic CD8+ T lymphocytes (CTLs), natural killer (NK) cells, dendritic cells (DCs), and various negative immune regulatory cells, including regulatory T cells (Tregs), helper T cells (Th), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) [35]. The roles in tumor killing and immune suppression are shown in Figure 2.

Figure 2: 
Immune cells in the TME after drug action. In the early stages of tumorigenesis, the immune system aims to destroy cancer cells, but as the tumor develops and the drugs work, fibroblasts, macrophages, T cells and B cells play the opposite role. For example, CAFs can be activated to secrete angiogenic factors, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), under hypoxia and metabolic reprogramming; TAMs in the TME mostly tend to be M2-type TAMs; it can further promote tumor invasion by secreting MMP9 to support angiogenesis and invasion. TAMs are also able to inhibit Th1 immune responses by secreting CCL17, CCL18, CCL12, TGF-1, PGE2, and Il-10, protecting tumor cells from immune attack. In addition, TAMs are located around new blood vessels and can help cancer cells cross the blood vessel barrier. The figure was set up using BioRender.com.
Figure 2:

Immune cells in the TME after drug action. In the early stages of tumorigenesis, the immune system aims to destroy cancer cells, but as the tumor develops and the drugs work, fibroblasts, macrophages, T cells and B cells play the opposite role. For example, CAFs can be activated to secrete angiogenic factors, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), under hypoxia and metabolic reprogramming; TAMs in the TME mostly tend to be M2-type TAMs; it can further promote tumor invasion by secreting MMP9 to support angiogenesis and invasion. TAMs are also able to inhibit Th1 immune responses by secreting CCL17, CCL18, CCL12, TGF-1, PGE2, and Il-10, protecting tumor cells from immune attack. In addition, TAMs are located around new blood vessels and can help cancer cells cross the blood vessel barrier. The figure was set up using BioRender.com.

During tumorigenesis, tumor tissue can recruit monocytes to its vicinity and transform them into TAMs [36]. In general, it is the division of activated macrophages are divided into M1 and M2 subpopulations. M1 macrophages secrete pro-inflammatory molecules such as interleukin (IL)-1, IL-12, IL-18 and IL-23, tumor necrosis factor (TNF) and chemokine C-C pattern ligand (CCL). On the other hand, M2 macrophages are primarily induced by Th2 cells and play a role in wound healing and allergic responses. They exhibit pro-tumorigenic activity and are able to downregulate the immunostimulatory factor IL-12 and upregulate the immunosuppressive factor IL-10 [37, 38]. During cancer treatment, TAMs exhibit a highly immunosuppressive M2 profile, which induces MDR through multiple mechanisms [39]. First, TAMs possess the ability to produce various cytokines that support angiogenesis, including matrix metalloproteinase (MMP) 9, MMP13, and vascular endothelial growth factor (VEGF) [40, 41]. It has been reported that mice treated with doxorubicin for breast cancer found that doxorubicin-induced tumor cell fruit TAMs secreting MMP9, which modulates vascular architecture and reduces vascular leakage by acting on substrates in TME, thereby impairing the conductive effects of doxorubicin [42]. Secondly, TAMs produce transforming growth factor (TGF)-1, prostaglandin E2 (PGE2), and IL-10, which defend tumor cells against immune attack. Production of monocyte pneumogastrin-1 (MCP-1) and interleukin 10 (IL-10) has been reported to be stimulated by TGF-1 and mediated by cross-talk between Smad, PI3K/AKT, and BRAF-MAPK signaling pathways. TGF-β1 regulates phosphorylation of Smad3 and expression of MCP-1 and IL-10 but is inhibited by P144 (a TG-β1 blocking peptide). This evidence suggests that TAMs can produce immunosuppressive effects [43]. In addition, IL-10 may activate STAT3 and enhance the expression of the B-cell lymphoma 2 (Bcl-2) gene, which can have a negative impact on tumor prognosis. TAMs may cause drug resistance in breast cancer through the IL10/STAT3/Bcl-2 signaling pathway [44]. In addition, TAMs produce prostaglandin E2, indoleamine pyrrole-2 inducer, CCL17, CCL18 and CCL22, which inhibit Th1 immune responses [45], [46], [47]. Based on micropore assays, immunocompromised mice developing melanomas co-express high levels of CCL17/22 with M2-type TAMs. Combining CD40 mAb with CSF-1R inhibitors slows tumor development, reduces the prevalence of M2-type TAMs, and increases T-cell activation by inducing the release of TNF, IL-6, and IL-12 [48]. Finally, TAMs may also release histone proteases that activate NF-κB Alternatively, they may activate IL6 and STAT3, which can lead to tumor cell-mediated resistance [49, 50].

Immature heterogeneous cells called MDSCs, which are produced in the bone marrow, may significantly impair the responsiveness of immune cells [51]. By producing nitric oxide synthase-2 (NOS-2), which reduces cysteine required for T cell activation, and arginase (ARG-1), which interferes with T cell protein synthesis, which requires arginine, MDSCs directly inhibit CD8+ T cells [52, 53]. Additionally, MDSCs can secrete TGF and VEGF, promoting angiogenesis and severely reducing the activity of effector T cells and NK cells, while activating T regulatory cells (Treg) [54, 55]. In tumor-bearing mice, PDE5 inhibitors have been shown to block MDSCs-mediated immunosuppressive mechanisms. As T cells activate the release of IFN-, MDSCs are receptive to IL-13 and produce their specific IFN-γ. By interacting with each other, these two cytokines activate the ARG1 and NOS-2 enzymes, leading to the release of nitric oxide (NO) and reactive oxygen species (ROS). This process generates nitrite and initiates the apoptosis of CD8+ T cells. PDE5 inhibitors increase the levels of cyclic guanosine monophosphate, which reduces the concentration of cell membrane Ca2+ concentration, leading to a decrease in calcium-dependent protein kinase C activity, which prevents upregulation of IL4Rα, which in turn impairs the IL4Rα-ARG1 pathway and allows immunosuppression to be alleviated [56]. Tumor-bearing mice with functionally suppressed MDSCs exhibit increased CD8+ T cell responsiveness, suggesting that these cells play an important role in tumor-induced immunosuppression [57].

Tregs, a subpopulation of CD4+ T cells expressing Foxp3, suppress the anti-tumor response of the immune system [58]. miR-142-3p reduced adenylate cycle 9 (AC9) expression was inhibited by Foxp3, whereas cAMP was promoted by AC9. Elevated cAMP levels inhibit antigen-presenting cells (APCs) and effector T cells through gap-functioning intercellular communication [59]. Tregs can promote immunosuppression of TME by secreting executive and proenzyme B and the immunosuppressive cytokines IL-10, IL-35 and TGF-β or by inhibiting T cell activity through direct contact [35]. By considering the effect of iNOS inhibition on Treg production in mouse and human CD4+ T cells, TGF1-dependent Treg activation was ameliorated in the work of Jayaraman et al. [60]. Higher numbers of Foxp3+ Treg were observed in tumor-bearing mice and within tumors. The combination of iNOS inhibitors and cyclophosphamide effectively inhibits MDSCs and Tregs, accelerating the metastatic process and thereby inhibiting tumor progression in an immune-dependent manner [60].

The immune system plays a crucial role in immunosurveillance as immune cells from both the adaptive and innate immune systems infiltrate the TME to regulate tumor progression. The immune regulatory cells mentioned above, including TAMs, MDSCs and Tregs, can all influence tumor progression. During drug treatment, all of these cells can respond with a corresponding regulatory response, thereby diminishing the therapeutic effect of the drug and consequently leading to the occurrence of MDR.

Soluble factors in the TME act in a paracrine manner

Following medication, damage to the TME’s DNA may trigger a reaction to stress in neoplastic and tumor-associated stomatal cells, which causes the creation of a variety of soluble substances, including GM-CSF, G-CSF, SCF, VEGF, and IL3, which promote myelopoiesis and contribute, in part, to a blockade of myeloid cell maturation [61], and amphiregulin (AREG) and epidermal growth factor (EGF) aid in epithelial cell proliferation, which also encourages the invasive WNT families, promotes tumor growth and survival, and results in increased resistance to chemotherapeutic drugs, and IL-1, IL-6, IL-8, and IL-27, which control inflammatory cells activity, and epithelial cell proliferation is aided by AREG and EGF [62, 63]. This DNA damage response stops cell growth and inhibits tumor development in a cell-autonomous, intrinsic, autocrine, or paracrine manner. Study shows that lymphoma survival following the use of a mouse model of Burkitt’s lymphoma is influenced by the paracrine component of TME. After DNA damage, IL-6 is secreted and released in a p38 MAP kinase-dependent manner, exerting a pro-survival benefit by inducing Bcl2 family members. Blocking the inhibitory effect of Bcl2, IL-6’s ability to promote doxorubicin was inhibited [64]. Additionally, it has been shown that after DNA damage, nuclear factor control of the NF-κB gene enhancer for light polypeptide activates WNT16B, which then sends a paracrine signal to tumor cells to activate the classical WNT program [65]. WNT16B expression in the prostate of TME reduces treatment efficacy and accelerates tumor growth. Cyclic chemotherapy could enhance resistance to subsequent treatment through this mechanism, contributing to the non-autonomous role of cells in TME [65].

The TME promotes metabolic reprogramming

TME is chronically hypoxic due to rapid tumor expansion, increased metabolic levels, and relatively inadequate oxygen supply, which is directly related to tumor resistance [66], [67], [68], [69]. Hypoxia-inducible factor-1 (HIF-1) binds to hypoxia-responsive elements (HRE) in the nucleus and promotes genes for a number of metabolic pathways as well as angiogenesis, growth factor, and receptor release, glycolysis, and other cellular processes [70, 71]. In colorectal cancer, it is reported that HIF-1 may improperly activate the PI3K/AKT and Wnt/β-catenin signaling pathways, leading to tumor cells’ drug resistance to 5-fluorouracil [72]. The MDR1 gene encodes P-glycoprotein, an efflux drug transport protein, and can pump hydrophobic anticancer medicines into the tumor’s outer ring while preventing drug buildup in the tumor cells, thus affecting the effective concentration of drugs in cells, resulting in the occurrence of tumor MDR [73]. Hypoxia regulates P-glycoprotein, which is a HIF-1 downstream target. P-glycoprotein expression may be decreased by HIF-1 inhibition [74, 75]. When P-glycoprotein was inhibited in hypoxia by the combination of the selective P-glycoprotein efflux activity inhibitor, tariquidar, and proteasome inhibitors, multiple myeloma cells were re-sensitized to bortezomib and carfilzomib [76]. Additionally, hypoxia particularly causes tumor cells to produce VEGF, which produces antiapoptotic proteins (including survivin and Bcl-2) and stimulates the PI3K/Akt antiapoptotic pathway [77], [78], [79]. The activation of HIF-1 and NF-κB, the promotion of CCL28 and cytokine production, and the release of VEGF are other ways that hypoxia may trigger inflammation [80]. Due to the production of VEGF, patients with lung adenocarcinoma had considerably greater blood concentrations of CCL28 than healthy individuals [80].

In addition to increased oxygen demand, there is usually an abnormal increase in glucose and glutamine requirements in tumor cells. The Warburg effect explains how tumor cells readjust their metabolism to cope with a nutrient-poor environment and support the growth of malignant tumors; even when sufficient oxygen is available, tumor cells abnormally activate glycolysis and inhibit mitochondrial respiration [8182]. The final product of glycolysis is lactic acid, which is transported by cells through transporters. HIF-1α can participate in the transport process by regulating monocarboxylic acid transporter 4, causing the TME to become acidic [83]. The low pH of TME (pH 6.5–6.9) promotes extracellular matrix (ECM) degradation via MMP and tissue proteases, releasing VEGF to increase angiogenesis and inhibit the tumor antigen-induced immune response, which can lead to a tumor-invasive phenotype and chemotherapy resistance [84, 85]. In addition, hypoxia and acidic TME increase the degree of dissociation of weakly basic drugs (e.g., adriamycin) and increase protonation, reducing ion trapping and drug uptake, leading to multidrug resistance [85]. Transport by the proton pump enhances the lysosome’s extracellular acidity and intracellular acidity. Under acidic conditions, the ability of the lysosome to chelate weakly alkaline drugs is enhanced and the drug is trapped in the lysosomal lumen, unable to interact with the target, leading to cellular resistance [86].

TME usually has an acidic environment, which is due to the abnormal metabolism of tumor cells, which produces a large amount of lactic acid and other acidic metabolites, leading to a decrease in pH. This acidic environment is conducive to the proliferation, invasion and metastasis of tumor cells, and at the same time can inhibit the activity of immune cells, resulting in a decrease in the ability of the immune system to attack tumor cells [87, 88]. The supply of oxygen in the TME is often uneven, with areas of hypoxia. This is due to the underdevelopment of the tumor vasculature, resulting in an inadequate supply of oxygen. Hypoxia can induce tumor cells to produce adaptive responses such as enhanced glycolysis, activation of the gluconeogenic pathway, and apoptosis and necrosis. Hypoxia can also promote malignant phenotypic transformation of tumor cells and increase their invasiveness and drug resistance. In addition, a variety of enzymes are present in TME, including protein hydrolases and oxidoreductases, which play important roles in tumor cell invasion, metastasis and immune escape. For example, protein hydrolases can break down the extracellular matrix and basement membrane, helping tumor cells to cross the tissue barrier. Oxidoreductase, on the other hand, is involved in the regulation of oxidative stress and protects tumor cells from oxidative damage [89], [90], [91]. Hypoxic and acidic niches are symbiotic and interactive, and targeting alone cannot effectively inhibit MDR. Therefore, further exploration of therapeutic strategies and specific molecular mechanisms should be part of the key research directions for TME.

The TME affects drug absorption

Drug distribution within tumors mainly occurs through diffusion, and the drug permeation barrier is one of the most significant factors that hinder drug uptake. Of the components of the TME, the ECM, a fibrous network of extracellular proteins and other macromolecules secreted by all the cells in the TME, is the most important; these components include collagen, fibronectin, proteoglycans, elastin, and integrins [92]. In normal tissue, the ECM is loose and consists of collagen, elastin, and glycoproteins, which can provide stable tissue structure and the circumstances required for cell division and proliferation. In most solid tumors, however, the ECM consists of another form of connective tissue fiber that becomes denser and stiffer. In addition, TME increases collagen synthesis, promotes cross-linking with elastin, and enhances the deposition of ECM, resulting in increased tumor stiffness [93], and proteoglycans combine with collagen to form cartilage components, both of which contribute to the stiffness and firmness of the tumor tissue [94]. It overabundantly surrounds tumor cells with this stiff, thick ECM, which serves as a barrier to keep medications out of the cells. However, since blood and lymphatic arteries are often absent from thick ECM, the transport of oxygen, nutrients, and metabolites is hampered. Pathways by which hypoxia and increased metabolic load trigger antiapoptotic and drug resistance mechanisms [95]. The production of crosslinks between collagen molecules and elastin networks may be facilitated by five families of lysine oxidase isozymes (LOX and LOXL1-4). Therefore, an increase in lysine oxidase can affect drug distribution [96, 97]. It is possible to simulate hypoxia and the tumor microenvironment in a way that uses a 3D in vitro cellular model in order to study tumor immune escape mechanisms and demonstrate its impressive potential in drug discovery. The model successfully simulated the survival and adaptability of tumor cells under hypoxic conditions and their interactions with the immune system [98]. Inhibition of lysine oxidase can improve oxygenation, as discovered in research on colorectal, ovarian, and breast cancer [96]. By oxidizing the extracellular domain of the receptor for platelet-derived growth factor, several investigations have shown that lysine oxidase may directly influence the production of vascular endothelial growth factor A (VEGF-A) [99].

TAFs are one of the most critical components of TME and are essential for altering ECM. There are several sources of TAFs, including local fibroblasts, stellate cells, bone marrow mesenchymal stem cells, and cells derived from adipose tissue [100]. Although TAFs exhibit significant heterogeneity, they share the same biological functions: invasiveness, proliferation, and promotion of immune responses. TAFs can promote MDR through a variety of mechanisms [94, 101, 102]. This section mainly describes how TAFs interfere with tumor therapy by participating in ECM remodeling.

The production of a range of matrix metalloproteinases (MMPs), including MMP-1 and MMP-3, as well as the secretion of several matrix proteins, including fibronectin and type I collagen, by TAFs may contribute to degrade the normal ECM structure and stiffen the matrix. TAFs can secrete proteoglycan growth factors, promote ECM remodeling, participate in the formation of therapeutic barriers through interactions with integrin and cadherin, and hinder antineoplastic drugs and immune cells in clinical treatment [103, 104]. Hyaluronic acid (HA) causes interstitial fluid pressure to be high which contributes to extracellular matrix remodeling, reducing drug exchange through capillaries [105]. In a TGF-dependent manner, TAFs can facilitate HA synthesis by promoting EGF production, which then triggers the ERK-MAPK pathway linked to EGFR [106], [107], [108]. According to some studies, TAFs inhibit the ability of cancer cells to respond to chemotherapy by activating interleukin-1 receptor-associated kinase 4 (IRAK4), promoting NF-κB activity in the stroma, and additionally can affect the transport of the gemcitabine drug in a mouse model of pancreatic cancer [109], [110], [111]. In addition, pro-epidermal growth factor, hepatocyte growth factor (HGF), and stromal cell-derived factor-1 (Sdf-1) are just a few of the cytokines that TAFs may release, which activate a series of cascade reactions and eventually lead to MDR and tumor recurrence [112]. In lung cancer, TAFs secrete IL-6, IL-8, and HGF and activate the miR-21 and MEK/ERK pathways, which promotes resistance to EGFR tyrosine kinase inhibitors [113]. Sphingosine-1-phosphate (S1P) and prostaglandin E2, which result from COX-2 activation and are secreted by TAFs as well, operate either autocrine or paracrine to promote apoptosis and chemical resistance via PI3K-Akt/PKB pathway activation [114].

Another important factor affecting drug absorption is angiogenesis around the tumor. The design and purpose of the newly formed blood arteries at the tumor location were found to be abnormal, and the blood flow was blocked substantially [115]. The tumor vasculature is also highly permeable and leaky, favoring non-specific extravasation of blood components into the surrounding interstitial space. As a result, the TME is not only hypoxic and acidic but also surrounded by high interstitial pressure, which acts as a pathological barrier, preventing drugs from entering the tumor and leading to MDR [116]. On the other hand, leaking blood vessels can promote the extravasation of the drug in the tumor area, resulting in an increased concentration of the drug in the tumor, leading to an enhanced therapeutic effect, but accelerating the formation of MDR. Drug distribution is also affected by blood vessels’ aberrant shape and function. Due to the obstruction of blood flow, the drug concentration in contact with tumor cells makes it difficult to produce cytotoxicity [117]. And the insufficient supply of nutrients leads to difficulties in excreting metabolites, affecting drug efficacy [117]. In addition to the secretion of VEGF, vascular endothelial cells may also have an impact on the expression of adhesion molecules on immune cells and endothelial cells, such as the integrin ligand intercellular adhesion molecule 1 (ICAM 1) and vascular cell adhesion protein 1 (VCAM 1). An immune checkpoint protein called programmed cell death 1 ligand 1 (PD-L1) binds to PD-1 expressed on T cells, inhibiting its anti-cancer activity, possibly produced by tumor-associated production, and influencing resistance to tumor therapy by inhibiting anti-tumor capacity [118]. In Zhang et al.’s study [119], the effects of VEGF overexpression and VEGF receptor 2 inhibition on the response to doxorubicin in soft-tissue sarcoma were studied in immunodeficient mice. The outcomes demonstrated a high degree of vascular proliferation, a shorter incubation time, quicker development, greater chemical resistance, and a higher incidence of lung metastasis in the xenografts with high VEGF expression. Treatment with the anti-VEGFR2 monoclonal antibody resulted in a reduction in the number of microvessels and an increase in the vascular maturation index, which also facilitated the response to doxorubicin and inhibited VEGFR2 signaling. The above evidence suggests that chemoresistance in soft-tissue sarcomas is caused by the interaction between tumor-associated endothelium cells. on the other hand, antiangiogenic therapies such as bevacizumab can lead to a further decrease in the vasculature, conferring hypoxia and physical seclusion from circulating drugs [120].

Angiogenesis, the creation of formation new blood vessels from pre-existing ones, plays an important role in tumorigenesis. Benign tumor cells exist in a dormant state and are affected when they have difficulty obtaining an adequate blood supply. However, an ‘angiogenic switch’ occurs when dormant tumor cell angiogenesis is activated, and factors are secreted that induce endothelial cells to germinate and converge into tumor masses. In the hypoxic environment of the internal tumor mass, the dimeric protein complex of HIF-1 remains stable and activates the expression of numerous genes that contribute to the angiogenic process, including vascular VEGF, which promotes vascular permeability, and bFGF, which promotes endothelial cell growth [121, 122].

Thus, the microenvironment itself is not always an active ‘soil’ per se, but occurs only incidentally in the vicinity of tumor multiplication, and tumor angiogenesis is a complex process of pro- and anti-angiogenic signaling pathways generated by both malignant and non-malignant cells through autocrine and paracrine production. Vascular density can have both positive and negative effects on drug delivery and efficacy.

Interaction between the TME and cancer stem cells

A limited subpopulation of cancer cells known as CSCs has an extensive capacity for self-renewal and division, which may contribute to the development and spread of malignant tumors [123]. They resemble embryonic stem cells [123]. In colorectal CSCs, HIF-1 has been shown to work in conjunction with the TAFs paracrine signal TGF-2 to activate the hedgehog transcription factor GLI2. This enhances dry/de-differentiation and genetic resistance to chemotherapy [124]. CSCs themselves are insensitive to conventional treatments such as radiotherapy and chemotherapy, showing drug resistance characteristics. In addition, MDR cells may develop resistance to cytotoxic compounds to which they have never been exposed. MDR may involve a variety of biochemical mechanisms [125, 126]; the most common MDR phenotype, also referred to as classic MDR, is associated with overexpression of transmembrane proteins that membrane proteins extrude substrates in a unidirectional manner, reducing the intracellular concentration of cytotoxic drugs below the effective dose. In MDR tumor cells, most of the overexpressed transporter proteins belong to the ATP-binding cassette (ABC) family of transporters, which actively transport substrates, including chemotherapeutic drugs, against a concentration gradient using energy generated by ATP hydrolysis [127]. Indeed, overexpression of these proteins is one of the hallmarks of drug resistance.

ABC transporters, which may move chemotherapy medications out of cells, lower intracellular drug concentrations, and lessen the risk of drug-induced cell toxicity, are extensively expressed by CSCs [128, 129]. Chemotherapeutic drugs primarily act on DNA or its replication process, destroying proliferating tumor cells by blocking the production of DNA and RNA, adding alkyl groups, or inhibiting enzymes. However, most CSCs are in the G0 phase and do not produce DNA or undergo cell division, which allows them to escape the killing effect of chemotherapeutic drugs and survive [130]. The damage repair ability of CSCs is robust. Even after an injury, p53, ATM-Chk2, ATR-Chk1 DNA, and other damage response pathways can be activated, leading to cell cycle arrest in G1, S, and G2 phases. This allows sufficient time and opportunity for the cells to undergo repairs [131]. The maintenance of the stem-like characteristics of CSCs is the main cause of drug resistance.

As a result of sustained external stimulation, the TME is altered and CSCs are enriched, leading to more resistant tumor tissue. The TME has a significant hypoxic component, which is essential to maintain CSC stemness. In a hypoxic environment, numerous signaling pathways mediated by HIF-1 are activated and involved in various aspects of cellular metabolism, leading to drug resistance. In human glioma cells, HIF-1 α is upregulated in CD133-positive CSCs, promoting their ability to self-renew. Hypoxia-mediated growth of CD144-positive CSCs is prevented after HIF-1 knockdown. Hypoxia-driven CD133 amplification was decreased when the PI3K-AKT or ERK1/2 pathways were inhibited, indicating that these signaling pathways may regulate hypoxia responses. Finally, CSCs cultured under hypoxia preserved an undifferentiated phenotype, unlike those cultured under normoxia. These findings imply that CSCs respond to hypoxia by activating HIF-1 to enhance the self-renewal capacity of CD133-positive cells and prevent the differentiation of CSCs [132]. Hypoxia also maintains the quiescent state of CSCs, inhibits apoptosis, and makes cells resistant to the killing effect of the drug. In non-small cell lung cancer (NSCLC), the CSC population can be reduced by modulating the PI3K/AKT pathway to increase the anticancer effect of gefitinib and reverse drug resistance [133]. In addition, TAFs and M2 macrophages in TME can secrete different cytokines and growth factors, including VEGF, SDF-1, IL10, and IL7, which can regulate the proliferation of CSCs and maintain their populations. Vermeulen et al. [134] found that HGF secreted by TAFs activates the Wnt signaling pathway through its interaction with c-Met. This activation leads to the development of stem-like properties in tumor cells located at the periphery of colon cancer. Consequently, these cells become resistant to medication and contribute to the recurrence of colon cancer. In breast cancer, continuous NF-κB activation and prolonged P65 nuclear retention are required to maintain CD10GPR77 TAFs’ paracrine IL6/IL8 secretion function and to provide a survival niche for CSCs; to provide a suitable survival microenvironment for CSCs, promote the development of tumor and drug resistance [135]. In addition, tumor cells can also secrete IL-6, which promotes the secretion of large amounts of CXCL7 by mesenchymal stem cells, and CXCL7 can promote the secretion of IL-6 by tumor cells, thus creating a cycle; in this process, both tumor and mesenchymal stem cells produce many cytokines, including CXCL1, CXCL5, CXCL6, IL-8, and IL-6, which promote the self-replication capacity of tumor stem cells [136].

Targeting TME to overcome MDR

Numerous anti-tumor drugs target TME because it is the external ‘soil’ in which tumor cells thrive, including the use of CSF receptor antibodies to deplete TAMs, the development of drugs that remain active under hypoxic conditions, and the inhibition of P-glycoprotein expression. However, there is a trend towards resistance to TME therapy. The cellular and non-cellular components of TME work together to form a complex system of protection and repair that favors damaged tumor cells to alter their original biological properties and thus resist single targeting agents. Combination therapy is therefore a more reasonable strategy to overcome MDR against TME. We next summarize the diagnosis and treatment strategies for targeting the TME to overcome MDR, as shown in Supplementary Table 1.

Targeting the immune system of the TME

Normally, T cells use the tumor’s immune circuit to recognize and clear tumor cells from the body, but tumors use immune checkpoints, regulatory molecules that suppress the immune system, to create an immune escape [137]. Immune checkpoint blockade (ICB) aids immune cells in their anti-tumor actions by designing relevant antibodies to inhibit immune escape. It has been documented that one of how immune escape occurs is through the interaction of the programmed apoptotic protein PD-1 with the ligands PD-L1 or PD-L2, thereby preventing T-cell activation [138]. Thus, in TME, the binding of PD-1 to ligands may be reduced by overexpression of PD-L1 or PD-L2, or by PD-1 inhibitors (e.g., pembrolizumab), thereby suppressing the intratumoral immune response and promoting T-cell activation [139]. In addition, anthracyclines, cyclophosphamide, oxaliplatin, and statins can induce tumor immunogenic cell death (ICD), release DAMPs and stimulate immune responses via APC effector T cells (e.g., DCs) [140]. To date, numerous studies have shown that the combination of chemotherapy and ICB is more effective. For example, pembrolizumab in combination with chemotherapy (NCT02039674) is the first-line therapy for metastatic NSCLC, with response rates rising by nearly half compared to monotherapy and the likelihood of disease progression reduced by nearly half [141]. Pembrolizumab in combination with pemetrexed and platinum is the first-line therapy for tumors lacking EGFR and ALK gene mutations in patients with metastatic NSCLC [142]. Pembrolizumab in combination with the conventional chemotherapeutic agents carboplatin, paclitaxel and paclitaxel, whose approval was based on the results of the phase III clinical study KEYNOTE-355 (NCT02819518), for the treatment of incurable, locally recurrent or spread triple-negative breast cancer, reducing the risk of disease progression and death by 35 % [143].

In addition, studies have shown that the combination of VEGF and ICB is effective because immune cells are unable to penetrate the tumor due to their vascularity and ECM. Atezolizumab, a PD-L1 monoclonal antibody, in combination with bevacizumab and chemotherapy can be effective in treating metastatic non-squamous NSCLC, but in patients without EGFR or ALK mutations [144]. Meanwhile, bevacizumab combined with TH-302, a drug activated in hypoxia, can be used for clinical effect in treatment-resistant glioblastoma multiforme, according to early findings from a phase II clinical study (NCT02342379) [145]. First-line treatments for advanced renal cell carcinoma (RCC) including tyrosine kinase inhibitors or cytokine therapy have failed. Fortunately, according to the results of the phase III KEYNOTE-426 study (NCT02853331), pembrolizumab combined with a tyrosine kinase inhibitor (axitinib) could be used as first-line treatment in advanced RCC, superior to sunitinib, and its combination saved almost half of the patients who died [146]. Meanwhile, results from the phase III JAVELIN Renal 100 trial (NCT02684006) suggest that the PD-L1 monoclonal antibody album in combination with axitinib could be used as a first course of treatment for patients with advanced RCC [147]. Further, pembrolizumab in combination with lenvatinib treatment was effective in controlling renal, endometrial and hepatocellular carcinoma, with control rates of 83–100 % for these three diseases [148]. The study found that the use of a new drug targeting the DNA repair pathway AZD6738, an ATR inhibitor [149]. And combining tazemetostat with other treatments may work better in treating patients with hematological malignancies and may improve disease response and durability of response. In addition, the study is to assess the safety and tolerability, obtain maximum tolerated dose (MTD) and/or the recommended phase 2 dose (RP2D) of LM-101 as a single agent or in combination in patients with advanced malignant tumors.

In addition, another way in which immune escape can occur is that tumors can establish an immunosuppressive environment by producing ligands for immune checkpoint receptors, thereby limiting T cell activation [150], such as by relying on young plasmacytoid dendritic cells (pDCs) [151]. The literature suggests that the role of Tregs and MDSCs with immunosuppressive potential is supported by immature pDCs, which lack type I interferon expression [152]. In response to this phenomenon, the TLR9 agonist CMP-001 was developed to activate immature pDCs by binding to the TLR9 receptor, enabling them to produce large amounts of type I interferon while promoting the production of other co-stimulants and presenting to T-cell tumor antigens, producing an efficient anti-tumor T-cell response [153]. Phase Ib study (NCT02680184) Results showed that the combination of CMP-001 and pembrolizumab for the treatment of patients with metastatic melanoma was significantly more effective than historical data on the efficacy of pembrolizumab alone, and a phase II clinical trial (NCT04698187) of the combination is therefore underway [154]. A phase I clinical study (NCT01885897) in which 19 % of patients with hematological malignancies had a clinical response to ALT-803 drug therapy with good treatment outcomes [155]. Phase Ib dose-escalation clinical study (NCT02523469) to assess the effectiveness of combination therapy with ALT-803 in combination with nivolumab in patients with NSCLC and safety and is currently in a phase II trial [156]. NKTR-214 and nivolumab are being used as first-line therapy in metastatic melanoma in PIVOT-02, a dose-escalation study (NCT03635983) that is investigating the safety and immunological efficacy of both agents with good tolerability, independent of baseline PD-L1 status [157].

Tumor-infiltrating lymphocyte (TIL) therapy has recently attracted a lot of interest due to the development of CAR-T therapy. TIL are TIL cells isolated from tissue near a tumor, expanded in vitro by the addition of the growth factor IL-2, and then infused back into the patient to amplify the immune response and treat primary or secondary tumors. TIL can kill primary or secondary tumors directly by releasing cytotoxic tumor cells. In addition, it can control the immune response and enhance the body’s ability to eliminate cancer cells [158]. In a multicentre phase II clinical trial, it was found that in patients with advanced cervical cancer, those who received TIL in addition to chemotherapy were more likely to respond to treatment [159]. Data from the study showed a response rate of 1 in 3 and a disease control rate of more than half for ICB treatment with innovative-01 in melanoma patients who had failed previous treatment [160]. This result suggests that TIL has the potential to overcome resistance to ICB.

Targeting other components of the TME

To improve the overall uptake and uniform distribution of drugs in tumor cells, researchers have developed drug delivery systems (DDS) for systemic and/or local drug therapy [161]. Assembled from carriers, antitumor drugs and drug efflux pump inhibitors, DDSs can achieve better therapeutic outcomes by cooperatively addressing different biological signaling pathways, allowing patients to receive low doses of chemotherapeutic drugs in combination. In addition, multiple drugs delivered together can target the same cellular pathway to maximize efficacy. Nanoparticle (NP) carriers prevent drug molecules from being recognized by efflux pumps, and intracellular drug release minimizes the rate of efflux-based drug transport in vitro [160]. Physical properties such as ECM and ECS in the tumor microenvironment can affect the diffusion and penetration ability of nanoparticles. The composition and structure of ECM and ECS in tumor tissues are more complex, including collagen, elastin, fibronectin, etc. Abnormal accumulation and distribution of these components may hinder the diffusion and penetration of nanoparticles. Secondly, chemical factors such as acidity, redox state and cytokines in the tumor microenvironment can also affect the drug delivery efficiency of nanoparticles [162], [163], [164], [165], [166], [167], [168]. For example, the acidic environment in the tumor tissue may affect the stability and drug-release behavior of nanoparticles, while cytokines may influence the interaction and uptake efficiency of nanoparticles with tumor cells. In addition, biological factors such as immune cells and stromal cells in the tumor microenvironment may also have an impact on the drug delivery of nanoparticles. For example, immune cells such as TAMs and DCs may affect the phagocytosis and processing of nanoparticles, which in turn may affect drug efficacy [169], [170], [171], [172], [173], [174]. Therefore, in recent years, researchers have increasingly investigated nanodrug delivery systems (NDDSs) [175]. NDDSs involve the direct delivery of drugs to the tumor, thereby reducing toxicity and bypassing many NDDSs including Doxil [176] and Abraxane [177]. However, hypoxia, low pH, and intercellular pressure in TME are important causes of MDR and can also lead to suboptimal therapeutic efficacy of foreign vector NDDSs. Therefore, there is a need to improve the oxygenation of the tumor. A study assessed the ability of CRLX101, an investigational nanoparticle-drug conjugate containing the payload camptothecin (CPT), to improve therapeutic responses as compared to standard chemotherapy [178]. EP0057 consists of a sugar molecule cyclodextrin linked to a chemotherapy drug called camptothecin. The combined molecule or “nanoparticle drug conjugate” travels through the blood. Once inside cancer cells, the chemotherapy drug is released from the molecule. Olaparib is a drug that may stop cancer cells from repairing the DNA damage caused by chemotherapy. Researchers describe the safety of EP0057 and olaparib, as well as evaluating the combination for the treatment of specific types of lung cancer in small cell lung cancer (SCLC) [179].

In addition, tumor-derived exosomes play an important role in the cancer immune microenvironment and cancer immunotherapy [180]. In terms of the immune microenvironment, tumor-derived exosomes can influence the function and status of a wide range of immune cells. For example, they can activate endothelial cells to support tumor angiogenesis and thrombosis, convert fibroblasts and bone marrow mesenchymal stem cells into myofibroblasts, and thereby promote tumor angiogenesis and metastasis. In addition, tumor-derived exosomes can create an immunosuppressive microenvironment by inducing apoptosis, impairing the function of effector T cells and NK cells, inhibiting DCs differentiation, expanding MDSCs and promoting Treg cell activity [181], [182], [183], [184]. In immunotherapy, exosomes can be used as targets for cancer immunotherapy. Several studies have identified that exosomes of tumor origin can carry tumor-associated molecular and genetic information that affects other cells and tissues in the tumor microenvironment, thereby influencing tumor development and metastasis. Therefore, the efficacy of existing immunotherapeutic strategies can be enhanced by interfering with the secretion and function of exosomes. For example, inhibiting exosome production can reduce the immune escape of tumor cells and improve the killing effect of immune cells; interfering with the function of exosomes can reduce the drug resistance of tumor cells and improve the efficacy of chemotherapeutic drugs [185], [186], [187].

Reconstruction of the vascular system and reprogramming of the ECM can be effective in alleviating the abnormal stress of TME [11, 188], [189], [190], [191], [192]. The approval of the anti-angiogenic drug bevacizumab for the treatment of metastatic colorectal cancer indicates confidence in this treatment modality [193]. In clinical practice, anti-angiogenic drugs are often used in combination with tyrosine kinase inhibitors to prevent MDR [164]. In addition, the combination of anti-angiogenic drugs and immune checkpoint inhibitors can partially alleviate immunosuppression and improve therapeutic efficiency [194]. Anti-angiogenic drugs can also increase the delivery of NDDS by inducing vascular normalization [195], [196], [197]. In addition to vascular systemic remodeling, ECM remodeling plays an important role in metabolic remodeling. Losartan, an angiotensin receptor inhibitor, has been shown to inhibit TGF-β signaling, target collagen and hyaluronic acid synthesis in the ECM, and slow fibrosis [198], [199], [200], [201]. A phase II clinical trial (NCT01821729) showed that the neoadjuvant chemotherapy regimen FOLFIRINOX combined with losartan reduced advanced pancreatic ductal carcinoma [202]. Other agents with anti-fibrotic properties, such as dexamethasone [203] and metformin, have also been used to restore ECM in TME [204, 205].

TAFs-based stromal cells and neovascular systems promote tumor progression and drug resistance. Therefore, reducing the impact of TAFs and reversing the distortion of the neovascular network is essential to overcome TME component-induced MDR. Cibucizumab, a fibroblast activation protein-α(FAP-α) specific antigen that targets the surface of TAFs, has been shown to maintain disease stability in patients with FAP α-positive tumors in phase I/II clinical studies, but the efficacy of monotherapy remains to be determined [206]. FAP and TAF DNA vaccines increased infiltration of CD8+ and CD4+ T cells into tumors and, in addition, targeting protein tyrosine kinase-2, which is involved in stromal fibrosis in mice, increased effector T cell infiltration and drug delivery, thereby enhancing immunotherapy and prolonging survival in mice [207]. Simlukafusp alfa (FAP-IL2v. RO6874281/RG7461) is an immunocytokine-containing antibody against FAP and a variant of IL2 that predisposes to IL2R βγ. The triple combination of FAP-IL2v with an anti-PD-L1 antibody and an agonistic CD40 antibody was most efficacious. These data indicate that FAP-IL2v is a potent immunocytokine that potentiates the efficacy of different T-cell and NK-cell-based cancer immunotherapies (NCT02627274 NCT03875079) [208].

Regardless of the method used, targeting TME may have a more pronounced and definitive modulatory effect on the MDR response than directly targeting tumor cells. The development and composition of TME are complex and variable, while the response and adaptation to various therapies remain unclear, and many non-tumor host cells with broad-spectrum functions are also present in TME. Our understanding of TME remains limited, which poses a challenge to elucidate the mechanisms of TME-mediated MDR, identify specific target cells and develop subsequent interventions.

Conclusions

Overall, we have focused on the importance of drug therapy in cancer treatment and the challenge of MDR of cancer cells to therapy (Figure 3). Available evidence suggests that the TME plays a crucial role in regulating tumour cell progression, metastasis, immune escape and drug resistance. Interactions between cancer cells and non-malignant cells alter the tumour microenvironment and promote drug resistance. The review focuses on the role of TME in promoting chemoresistance in tumour cells through a variety of mechanisms, including inhibition of immune clearance of tumour cells, stimulation of soluble paracrine factor release, metabolic reprogramming, and induction of changes in peritumour stomatal cells and blood vessels. Finally, the review also explores clinical therapeutic strategies against TME, providing insights and rationale for reversing multidrug resistance. Although there has been much research on the role of the TME in promoting tumor resistance, given the multi-layered and complex nature of TME, there is a need for further research into anti-MDR therapies that target TME molecules or signaling pathways. To date, targeting TME for the treatment of MDR cancers has shown positive therapeutic effects and offers potential reversal possibilities for patients with refractory or recurrent tumors. Therefore, a deeper exploration of the molecular mechanisms underlying the interaction between TME and MDR and the study of multi-targeted therapeutic agents to optimize diagnosis and treatment will offer new hope for tumor patients. Nonetheless, translating TME findings into clinical practice presents obstacles, and multidisciplinary collaboration and closer laboratory-clinical partnerships are needed to ensure that the reproducibility and reliability of TME findings are incorporated into clinical trials and treatment recommendations. These collaborative efforts will improve the translation of TME findings into clinical applications.

Figure 3: 
Determinants of cancer drug resistance and how to overcome them.
Figure 3:

Determinants of cancer drug resistance and how to overcome them.


Corresponding Author: Daiming Fan, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China; and State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, 710032, Shaanxi, China, E-mail:

Award Identifier / Grant number: 81822031

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: Material preparation, data collection, analysis, and writing were performed by Lingnan Meng and Ying Zheng. The figures and table of the manuscript were drawn by Lingnan Meng. Daiming Fan and Hao Liu commented on previous versions of the manuscript. All authors read and approved the final manuscript.

  4. Competing interests: The authors state no conflict of interest.

  5. Research funding: This research was funded by the National Natural Science Foundation of China (No. 81822031).

  6. Data availability: Not applicable.

References

1. Bray, F, Jemal, A, Grey, N, Ferlay, J, Forman, D. Global cancer transitions according to the Human Development Index (2008–2030): a population-based study. Lancet Oncol 2012;13:790–801. https://doi.org/10.1016/s1470-2045(12)70211-5.Search in Google Scholar PubMed

2. Sung, H, Ferlay, J, Siegel, RL, Laversanne, M, Soerjomataram, I, Jemal, A, et al.. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin 2021;71:209–49. https://doi.org/10.3322/caac.21660.Search in Google Scholar PubMed

3. Mattiuzzi, C, Lippi, G. Current cancer epidemiology. J Epidemiol Global Health 2019;9:217–22. https://doi.org/10.2991/jegh.k.191008.001.Search in Google Scholar PubMed PubMed Central

4. Igarashi, Y, Sasada, T. Cancer vaccines: toward the next breakthrough in cancer immunotherapy. J Immunol Res 2020;2020:5825401–13. https://doi.org/10.1155/2020/5825401.Search in Google Scholar PubMed PubMed Central

5. Slichenmyer, WJ, Von Hoff, DD. Taxol: a new and effective anti-cancer drug. Anti Cancer Drugs 1991;2:519–30. https://doi.org/10.1097/00001813-199112000-00002.Search in Google Scholar

6. Jasmin, C. Gene therapy and cancer: from concepts to clinical applications. Pathol Biol 1993;41:686–90.Search in Google Scholar

7. Lotz, JP, Pene, F, Bouleuc, C, Andre, T, Gligorov, J, Avenin, D, et al.. High-dose chemotherapy and autologous stem cell support for the treatment of gynecologic tumors in adults. Contracept Fertil Sex 1996;24:307–18.Search in Google Scholar

8. Guo, Q, Li, X, Cui, MN, Sun, JL, Ji, HY, Ni, BB, et al.. CD13: a key player in multidrug resistance in cancer chemotherapy. Oncol Res 2020;28:533–40. https://doi.org/10.3727/096504020x15919605976853.Search in Google Scholar PubMed PubMed Central

9. Solyanik, GI. Multifactorial nature of tumor drug resistance. Exp Oncol 2010;32:181–5.Search in Google Scholar

10. Assaraf, YG, Brozovic, A, Gonçalves, AC, Jurkovicova, D, Linē, A, Machuqueiro, M, et al.. The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist Updates: Rev Commen Antimicrob Anticancer Chemother 2019;46:100645. https://doi.org/10.1016/j.drup.2019.100645.Search in Google Scholar PubMed

11. Chen, Q, Liu, G, Liu, S, Su, H, Wang, Y, Li, J, et al.. Remodeling the tumor microenvironment with emerging nanotherapeutics. Trends Pharmacol Sci 2018;39:59–74. https://doi.org/10.1016/j.tips.2017.10.009.Search in Google Scholar PubMed

12. Arneth, B. Tumor microenvironment. Medicina (Kaunas, Lithuania) 2019;56:15. https://doi.org/10.3390/medicina56010015.Search in Google Scholar PubMed PubMed Central

13. Ni, Y, Zhou, X, Yang, J, Shi, H, Li, H, Zhao, X, et al.. The role of tumor-stroma interactions in drug resistance within tumor microenvironment. Front Cell Dev Biol 2021;9:637675. https://doi.org/10.3389/fcell.2021.637675.Search in Google Scholar PubMed PubMed Central

14. Chen, J, Zhu, H, Yin, Y, Jia, S, Luo, X. Colorectal cancer: metabolic interactions reshape the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2022;1877:188797. https://doi.org/10.1016/j.bbcan.2022.188797.Search in Google Scholar PubMed

15. Wang, H, Wang, B, Jiang, J, Wu, Y, Song, A, Wang, X, et al.. SnSe nanosheets mimic lactate dehydrogenase to reverse tumor acid microenvironment metabolism for enhancement of tumor therapy. Molecules 2022;27:8552. https://doi.org/10.3390/molecules27238552.Search in Google Scholar PubMed PubMed Central

16. Wang, L, Dunmall, LSC, Cheng, Z, Wang, Y. Remodeling the tumor microenvironment by oncolytic viruses: beyond oncolysis of tumor cells for cancer treatment. J Immunother Cancer 2022;10:e004167. https://doi.org/10.1136/jitc-2021-004167.Search in Google Scholar PubMed PubMed Central

17. Wang, L, Yang, JD, Yoo, CC, Lai, KKY, Braun, J, McGovern, DPB, et al.. Magnetic resonance imaging for characterization of hepatocellular carcinoma metabolism. Front Physiol 2022;13:1056511–1. https://doi.org/10.3389/fphys.2022.1056511.Search in Google Scholar PubMed PubMed Central

18. Wu, C, Mao, Y, Wang, X, Li, P, Tang, B. Deep-tissue fluorescence imaging study of reactive oxygen species in a tumor microenvironment. Anal Chem 2022;94:165–76. https://doi.org/10.1021/acs.analchem.1c03104.Search in Google Scholar PubMed

19. Zhu, Y, Li, X, Wang, L, Hong, X, Yang, J. Metabolic reprogramming and crosstalk of cancer-related fibroblasts and immune cells in the tumor microenvironment. Front Endocrinol 2022;13:13. https://doi.org/10.3389/fendo.2022.988295.Search in Google Scholar PubMed PubMed Central

20. Li, L, Tian, Y, Fu, H, Yao, Q, Wang, L, Lou, L. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023;13:161–75.10.1016/j.biopha.2023.114504Search in Google Scholar PubMed

21. Li, X, Wenes, M, Romero, P, Huang, SC-C, Fendt, S-M, Ho, P-C. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol 2019;16:425–41. https://doi.org/10.1038/s41571-019-0203-7.Search in Google Scholar PubMed

22. Li, T, Qiao, T. Unraveling tumor microenvironment of small-cell lung cancer: implications for immunotherapy. Semin Cancer Biol 2022;86:117–25. https://doi.org/10.1016/j.semcancer.2022.09.005.Search in Google Scholar PubMed

23. Castells, M, Thibault, B, Delord, JP, Couderc, B. Implication of tumor microenvironment in chemoresistance: tumor-associated stromal cells protect tumor cells from cell death. Int J Mol Sci 2012;13:9545–71. https://doi.org/10.3390/ijms13089545.Search in Google Scholar PubMed PubMed Central

24. Shoji, M, Hancock, WW, Abe, K, Micko, C, Casper, KA, Baine, RM, et al.. Activation of coagulation and angiogenesis in cancer – immunohistochemical localization in situ of clotting proteins and vascular endothelial growth factor in human cancer. Am J Pathol 1998;152:399–411.Search in Google Scholar

25. Leong, A. Total mesorectal excision (TME) – twenty years on. Ann Acad Med Singapore 2003;32:159–62. https://doi.org/10.47102/annals-acadmedsg.v32n2p159.Search in Google Scholar

26. Savitskiy, VP, Zorin, VP, Potapnev, MP. Accumulation of chlorine e6 derivatives in cells with different level of expression and function activity of multidrug resistance protein P-gp 170. Exp Oncol 2005;27:47–51.Search in Google Scholar

27. Xiang, R, Luo, Y, Niethammer, AG, Reisfeld, RA. Oral DNA vaccines target the tumor vasculature and microenvironment and suppress tumor growth and metastasis. Immunol Rev 2008;222:117–28. https://doi.org/10.1111/j.1600-065x.2008.00613.x.Search in Google Scholar PubMed

28. Karar, J, Maity, A. Modulating the tumor microenvironment to increase radiation responsiveness. Cancer Biol Ther 2009;8:1994–2001. https://doi.org/10.4161/cbt.8.21.9988.Search in Google Scholar PubMed PubMed Central

29. Pure, E. The road to integrative cancer therapies: emergence of a tumor-associated fibroblast protease as a potential therapeutic target in cancer. Expert Opin Ther Targets 2009;13:967–73. https://doi.org/10.1517/14728220903103841.Search in Google Scholar PubMed

30. Shinohara, ET, Maity, A. Increasing sensitivity to radiotherapy and chemotherapy by using novel biological agents that alter the tumor microenvironment. Curr Mol Med 2009;9:1034–45. https://doi.org/10.2174/156652409789839107.Search in Google Scholar PubMed

31. Leibovici, J, Itzhaki, O, Huszar, M, Sinai, J. Targeting the tumor microenvironment by immunotherapy: part 2. Immunotherapy 2011;3:1385–408. https://doi.org/10.2217/imt.11.112.Search in Google Scholar PubMed

32. Chi, K-H, Wang, Y-S, Kao, S-J. Improving radioresponse through modification of the tumor immunological microenvironment. Cancer Biother Rad 2012;27:6–11. https://doi.org/10.1089/cbr.2011.1048.Search in Google Scholar PubMed

33. Dalton, DK, Noelle, RJ. The roles of mast cells in anticancer immunity. Cancer Immunol Immunother 2012;61:1511–20. https://doi.org/10.1007/s00262-012-1246-0.Search in Google Scholar PubMed PubMed Central

34. El-Nikhely, N, Larzabal, L, Seeger, W, Calvo, A, Savai, R. Tumor-stromal interactions in lung cancer: novel candidate targets for therapeutic intervention. Expet Opin Invest Drugs 2012;21:1107–22. https://doi.org/10.1517/13543784.2012.693478.Search in Google Scholar PubMed

35. Sharma, P, Hu-Lieskovan, S, Wargo, JA, Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. https://doi.org/10.1016/j.cell.2017.01.017.Search in Google Scholar PubMed PubMed Central

36. Netea-Maier, RT, Smit, JWA, Netea, MG. Metabolic changes in tumor cells and tumor-associated macrophages: a mutual relationship. Cancer Lett 2018;413:102–9. https://doi.org/10.1016/j.canlet.2017.10.037.Search in Google Scholar PubMed

37. Noy, R, Pollard, JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity 2014;41:49–61. https://doi.org/10.1016/j.immuni.2014.06.010.Search in Google Scholar PubMed PubMed Central

38. Yunna, C, Mengru, H, Lei, W, Weidong, C. Macrophage M1/M2 polarization. Eur J Pharmacol 2020;877:173090. https://doi.org/10.1016/j.ejphar.2020.173090.Search in Google Scholar PubMed

39. Biswas, SK, Sica, A, Lewis, CE. Plasticity of macrophage function during tumor progression: regulation by distinct molecular mechanisms. J Immunol (Baltimore, Md: 1950) 2008;180:2011–7, https://doi.org/10.4049/jimmunol.180.4.2011.Search in Google Scholar PubMed

40. Yu, Q, Stamenkovic, I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Gene Dev 2000;14:163–76. https://doi.org/10.1101/gad.14.2.163.Search in Google Scholar

41. Larionova, I, Cherdyntseva, N, Liu, T, Patysheva, M, Rakina, M, Kzhyshkowska, J. Interaction of tumor-associated macrophages and cancer chemotherapy. OncoImmunology 2019;8:1596004. https://doi.org/10.1080/2162402x.2019.1596004.Search in Google Scholar

42. Nakasone, ES, Askautrud, HA, Kees, T, Park, J-H, Plaks, V, Ewald, AJ, et al.. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell 2012;21:488–503. https://doi.org/10.1016/j.ccr.2012.02.017.Search in Google Scholar PubMed PubMed Central

43. Diaz-Valdes, N, Basagoiti, M, Dotor, J, Aranda, F, Monreal, I, Ignacio Riezu-Boj, J, et al.. Induction of monocyte chemoattractant protein-1 and interleukin-10 by TGF beta 1 in melanoma enhances tumor infiltration and immunosuppression. Cancer Res 2011;71:812–21. https://doi.org/10.1158/0008-5472.can-10-2698.Search in Google Scholar

44. Yang, C, He, L, He, P, Liu, Y, Wang, W, He, Y, et al.. Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway. Med Oncol 2015;32:14. https://doi.org/10.1007/s12032-014-0352-6.Search in Google Scholar PubMed

45. Komohara, Y, Jinushi, M, Takeya, M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci 2014;105:1–8. https://doi.org/10.1111/cas.12314.Search in Google Scholar PubMed PubMed Central

46. Son, B, Lee, S, Youn, H, Kim, E, Kim, W, Youn, B. The role of tumor microenvironment in therapeutic resistance. Oncotarget 2017;8:3933–45. https://doi.org/10.18632/oncotarget.13907.Search in Google Scholar PubMed PubMed Central

47. Korbecki, J, Olbromski, M, Dziegiel, P. CCL18 in the progression of cancer. Int J Mol Sci 2020;21:7955. https://doi.org/10.3390/ijms21217955.Search in Google Scholar PubMed PubMed Central

48. Perry, CJ, Munoz-Rojas, AR, Meeth, KM, Kellman, LN, Amezquita, RA, Thakral, D, et al.. Myeloid-targeted immunotherapies act in synergy to induce inflammation and antitumor immunity. J Exp Med 2018;215:877–93. https://doi.org/10.1084/jem.20171435.Search in Google Scholar PubMed PubMed Central

49. Jinushi, M, Chiba, S, Yoshiyama, H, Masutomi, K, Kinoshita, I, Dosaka-Akita, H, et al.. Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc Natl Acad Sci USA 2011;108:12425–30. https://doi.org/10.1073/pnas.1106645108.Search in Google Scholar PubMed PubMed Central

50. Wang, H, Wang, X, Li, X, Fan, Y, Li, G, Guo, C, et al.. CD68(+)HLA-DR+ M1-like macrophages promote motility of HCC cells via NF-kappa B/FAK pathway. Cancer Lett 2014;345:91–9. https://doi.org/10.1016/j.canlet.2013.11.013.Search in Google Scholar PubMed

51. Talmadge, JE, Gabrilovich, DI. History of myeloid-derived suppressor cells. Nat Rev Cancer 2013;13:739–52. https://doi.org/10.1038/nrc3581.Search in Google Scholar PubMed PubMed Central

52. Bronte, V, Serafini, P, Mazzoni, A, Segal, DM, Zanovello, P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol 2003;24:302–6. https://doi.org/10.1016/s1471-4906(03)00132-7.Search in Google Scholar PubMed

53. Romano, A, Parrinello, NL, La Cava, P, Tibullo, D, Giallongo, C, Camiolo, G, et al.. PMN-MDSC and arginase are increased in myeloma and may contribute to resistance to therapy. Expert Rev Mol Diagn 2018;18:675–83. https://doi.org/10.1080/14737159.2018.1470929.Search in Google Scholar PubMed

54. Condamine, T, Ramachandran, I, Youn, JI, Gabrilovich, DI. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med 2015;66:97–110. https://doi.org/10.1146/annurev-med-051013-052304.Search in Google Scholar PubMed PubMed Central

55. Kumar, V, Patel, S, Tcyganov, E, Gabrilovich, DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol 2016;37:208–20. https://doi.org/10.1016/j.it.2016.01.004.Search in Google Scholar PubMed PubMed Central

56. Serafini, P, Meckel, K, Kelso, M, Noonan, K, Califano, J, Koch, W, et al.. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med 2006;203:2691–702. https://doi.org/10.1084/jem.20061104.Search in Google Scholar PubMed PubMed Central

57. Terabe, M, Matsui, S, Park, JM, Mamura, M, Noben-Trauth, N, Donaldson, DD, et al.. Transforming growth factor-beta production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. J Exp Med 2003;198:1741–52. https://doi.org/10.1084/jem.20022227.Search in Google Scholar PubMed PubMed Central

58. Liyanage, UK, Moore, TT, Joo, HG, Tanaka, Y, Herrmann, V, Doherty, G, et al.. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol (Baltimore, Md: 1950) 2002;169:2756–61, https://doi.org/10.4049/jimmunol.169.5.2756.Search in Google Scholar PubMed

59. Zhao, H, Liao, X, Kang, Y. Tregs: where we are and what comes next? Front Immunol 2017;8:8. https://doi.org/10.3389/fimmu.2017.01578.Search in Google Scholar PubMed PubMed Central

60. Jayaraman, P, Alfarano, MG, Svider, PF, Parikh, F, Lu, G, Kidwai, S, et al.. iNOS expression in CD4+ T cells limits Treg induction by repressing TGFβ1: combined iNOS inhibition and Treg depletion unmask endogenous antitumor immunity. Clin Cancer Res 2014;20:6439–51. https://doi.org/10.1158/1078-0432.ccr-13-3409.Search in Google Scholar

61. Gabrilovich, DI, Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009;9:162–74. https://doi.org/10.1038/nri2506.Search in Google Scholar PubMed PubMed Central

62. Sun, Y, Nelson, PS. Molecular pathways: involving microenvironment damage responses in cancer therapy resistance. Clin Cancer Res 2012;18:4019–25. https://doi.org/10.1158/1078-0432.ccr-11-0768.Search in Google Scholar

63. Gomez-Sarosi, L, Sun, Y, Coleman, I, Bianchi-Frias, D, Nelson, PS. DNA damage induces a secretory program in the quiescent TME that fosters adverse cancer phenotypes. Mol Cancer Res: MCR 2017;15:842–51. https://doi.org/10.1158/1541-7786.mcr-16-0387.Search in Google Scholar PubMed PubMed Central

64. Gilbert, LA, Hemann, MT. DNA damage-mediated induction of a chemoresistant niche. Cell 2010;143:355–66. https://doi.org/10.1016/j.cell.2010.09.043.Search in Google Scholar PubMed PubMed Central

65. Sun, Y, Campisi, J, Higano, C, Beer, TM, Porter, P, Coleman, I, et al.. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med 2012;18:1359–68. https://doi.org/10.1038/nm.2890.Search in Google Scholar PubMed PubMed Central

66. Najafi, M, Mortezaee, K, Majidpoor, J. Cancer stem cell (CSC) resistance drivers. Life Sci 2019;234:116781. https://doi.org/10.1016/j.lfs.2019.116781.Search in Google Scholar PubMed

67. Khalaf, K, Hana, D, Chou, JT-T, Singh, C, Mackiewicz, A, Kaczmarek, M. Aspects of the tumor microenvironment involved in immune resistance and drug resistance. Front Immunol 2021;12:12. https://doi.org/10.3389/fimmu.2021.656364.Search in Google Scholar PubMed PubMed Central

68. Liang, W, He, X, Bi, J, Hu, T, Sun, Y. Role of reactive oxygen species in tumors based on the ‘seed and soil’ theory: a complex interaction. Oncol Rep 2021;46:208. https://doi.org/10.3892/or.2021.8159.Search in Google Scholar PubMed PubMed Central

69. Weng, C-Y, Kao, C-X, Chang, T-S, Huang, Y-H. Immuno-metabolism: the role of cancer niche in immune checkpoint inhibitor resistance. Int J Mol Sci 2021;22:1258. https://doi.org/10.3390/ijms22031258.Search in Google Scholar PubMed PubMed Central

70. Hielscher, A, Gerecht, S. Hypoxia and free radicals: role in tumor progression and the use of engineering-based platforms to address these relationships. Free Radic Biol Med 2015;79:281–91. https://doi.org/10.1016/j.freeradbiomed.2014.09.015.Search in Google Scholar PubMed PubMed Central

71. Multhoff, G, Vaupel, P. Hypoxia compromises anti-cancer immune responses. Adv Exp Med Biol 2020;1232:131–43. https://doi.org/10.1007/978-3-030-34461-0_18.Search in Google Scholar PubMed

72. Dong, S, Liang, S, Cheng, Z, Zhang, X, Luo, L, Li, L, et al.. ROS/PI3K/Akt and Wnt/β-catenin signalings activate HIF-1α-induced metabolic reprogramming to impart 5-fluorouracil resistance in colorectal cancer. J Exp Clin Cancer Res 2022;41:15. https://doi.org/10.1186/s13046-021-02229-6.Search in Google Scholar PubMed PubMed Central

73. Sherlach, KS, Roepe, PD. Drug resistance associated membrane proteins. Front Physiol 2014;5:108. https://doi.org/10.3389/fphys.2014.00108.Search in Google Scholar PubMed PubMed Central

74. Comerford, KM, Wallace, TJ, Karhausen, J, Louis, NA, Montalto, MC, Colgan, SP. Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res 2002;62:3387–94.Search in Google Scholar

75. Chen, J, Ding, Z, Peng, Y, Pan, F, Li, J, Zou, L, et al.. HIF-1α inhibition reverses multidrug resistance in colon cancer cells via downregulation of MDR1/P-glycoprotein. PLoS One 2014;9:e98882. https://doi.org/10.1371/journal.pone.0098882.Search in Google Scholar PubMed PubMed Central

76. Muz, B, Kusdono, HD, Azab, F, de la Puente, P, Federico, C, Fiala, M, et al.. Tariquidar sensitizes multiple myeloma cells to proteasome inhibitors via reduction of hypoxia-induced P-gp-mediated drug resistance. Leuk Lymphoma 2017;58:2916–25. https://doi.org/10.1080/10428194.2017.1319052.Search in Google Scholar PubMed PubMed Central

77. Liu, Y, Cox, SR, Morita, T, Kourembanas, S. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells. Identification of a 5′ enhancer. Circ Res 1995;77:638–43. https://doi.org/10.1161/01.res.77.3.638.Search in Google Scholar PubMed

78. Nör, JE, Christensen, J, Mooney, DJ, Polverini, PJ. Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression. Am J Pathol 1999;154:375–84. https://doi.org/10.1016/s0002-9440(10)65284-4.Search in Google Scholar PubMed PubMed Central

79. Fukuda, S, Pelus, LM. Survivin, a cancer target with an emerging role in normal adult tissues. Mol Cancer Therapeut 2006;5:1087–98. https://doi.org/10.1158/1535-7163.mct-05-0375.Search in Google Scholar

80. Huang, G, Tao, L, Shen, S, Chen, L. Hypoxia induced CCL28 promotes angiogenesis in lung adenocarcinoma by targeting CCR3 on endothelial cells. Sci Rep 2016;6:27152. https://doi.org/10.1038/srep27152.Search in Google Scholar PubMed PubMed Central

81. Warburg, O. On the origin of cancer cells. Science 1956;123:309–14. https://doi.org/10.1126/science.123.3191.309.Search in Google Scholar PubMed

82. Vaupel, P, Multhoff, G. The warburg effect: historical dogma versus current rationale. Adv Exp Med Biol 2021;1269:169–77. https://doi.org/10.1007/978-3-030-48238-1_27.Search in Google Scholar PubMed

83. Ullah, MS, Davies, AJ, Halestrap, AP. The plasma membrane lactate transporter MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent mechanism. J Biol Chem 2006;281:9030–7. https://doi.org/10.1074/jbc.m511397200.Search in Google Scholar PubMed

84. Fischer, K, Hoffmann, P, Voelkl, S, Meidenbauer, N, Ammer, J, Edinger, M, et al.. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007;109:3812–9. https://doi.org/10.1182/blood-2006-07-035972.Search in Google Scholar PubMed

85. Taylor, S, Spugnini, EP, Assaraf, YG, Azzarito, T, Rauch, C, Fais, S. Microenvironment acidity as a major determinant of tumor chemoresistance: proton pump inhibitors (PPIs) as a novel therapeutic approach. Drug Resist Updates: Rev Comment Antimicrob Anticancer Chemother 2015;23:69–78. https://doi.org/10.1016/j.drup.2015.08.004.Search in Google Scholar PubMed

86. Zhitomirsky, B, Assaraf, YG. Lysosomal sequestration of hydrophobic weak base chemotherapeutics triggers lysosomal biogenesis and lysosome-dependent cancer multidrug resistance. Oncotarget 2015;6:1143–56. https://doi.org/10.18632/oncotarget.2732.Search in Google Scholar PubMed PubMed Central

87. Prasad, P, Gordijo, CR, Abbasi, AZ, Maeda, A, Ip, A, Rauth, AM, et al.. Multifunctional albumin-MnO2 nanoparticles modulate solid tumor microenvironment by attenuating hypoxia, acidosis, vascular endothelial growth factor and enhance radiation response. ACS Nano 2014;8:3202–12. https://doi.org/10.1021/nn405773r.Search in Google Scholar PubMed

88. Chen, Q, Feng, L, Liu, J, Zhu, W, Dong, Z, Wu, Y, et al.. Intelligent albumin-MnO2 nanoparticles as pH-/H2O2-responsive dissociable nanocarriers to modulate tumor hypoxia for effective combination therapy. Adv Mater 2016;28:7129–+. https://doi.org/10.1002/adma.201601902.Search in Google Scholar PubMed

89. Tang, Z, Zhang, H, Liu, Y, Ni, D, Zhang, H, Zhang, J, et al.. Antiferromagnetic pyrite as the tumor microenvironment-mediated nanoplatform for self-enhanced tumor imaging and therapy. Adv Mater 2017;29(47):1701683. https://doi.org/10.1002/adma.201701683.Search in Google Scholar PubMed

90. Gong, F, Yang, N, Wang, X, Zhao, Q, Chen, Q, Liu, Z, et al.. Tumor microenvironment-responsive intelligent nanoplatforms for cancer theranostics. Nano Today 2020;32:100851. https://doi.org/10.1016/j.nantod.2020.100851.Search in Google Scholar

91. Zhang, Y, Kim, I, Xu, Y, Yu, D-G, Song, W. Intelligent poly(L-histidine)-based nanovehicles for controlled drug delivery. J Contr Release 2022;349:963–82. https://doi.org/10.1016/j.jconrel.2022.08.005.Search in Google Scholar PubMed

92. Theocharis, AD, Manou, D, Karamanos, NK. The extracellular matrix as a multitasking player in disease. FEBS J 2019;286:2830–69. https://doi.org/10.1111/febs.14818.Search in Google Scholar PubMed

93. Xiong, G, Deng, L, Zhu, J, Rychahou, PG, Xu, R. Prolyl-4-hydroxylase α subunit 2 promotes breast cancer progression and metastasis by regulating collagen deposition. BMC Cancer 2014;14:1. https://doi.org/10.1186/1471-2407-14-1.Search in Google Scholar PubMed PubMed Central

94. Henke, E, Nandigama, R, Ergün, S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci 2019;6:160. https://doi.org/10.3389/fmolb.2019.00160.Search in Google Scholar PubMed PubMed Central

95. Jain, RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 2014;26:605–22. https://doi.org/10.1016/j.ccell.2014.10.006.Search in Google Scholar PubMed PubMed Central

96. Rossow, L, Veitl, S, Vorlová, S, Wax, JK, Kuhn, AE, Maltzahn, V, et al.. LOX-catalyzed collagen stabilization is a proximal cause for intrinsic resistance to chemotherapy. Oncogene 2018;37:4921–40. https://doi.org/10.1038/s41388-018-0320-2.Search in Google Scholar PubMed PubMed Central

97. Sutherland, RM, Eddy, HA, Bareham, B, Reich, K, Vanantwerp, D. Resistance to adriamycin in multicellular spheroids. Int J Radiat Oncol Biol Phys 1979;5:1225–30. https://doi.org/10.1016/0360-3016(79)90643-6.Search in Google Scholar PubMed

98. Ray, SK, Mukherjee, S. Imitating hypoxia and tumor microenvironment with immune evasion by employing three dimensional in vitro cellular models: impressive tool in drug discovery. Recent Pat Anti-Cancer Drug Discov 2022;17:80–91. https://doi.org/10.2174/1574892816666210728115605.Search in Google Scholar PubMed

99. Baker, AM, Bird, D, Welti, JC, Gourlaouen, M, Lang, G, Murray, GI, et al.. Lysyl oxidase plays a critical role in endothelial cell stimulation to drive tumor angiogenesis. Cancer Res 2013;73:583–94. https://doi.org/10.1158/0008-5472.can-12-2447.Search in Google Scholar

100. Itoh, G, Takagane, K, Fukushi, Y, Kuriyama, S, Umakoshi, M, Goto, A, et al.. Cancer-associated fibroblasts educate normal fibroblasts to facilitate cancer cell spreading and T-cell suppression. Mol Oncol 2022;16:166–87. https://doi.org/10.1002/1878-0261.13077.Search in Google Scholar PubMed PubMed Central

101. Ma, Y, Wang, Y, Xu, Z, Wang, Y, Fallon, JK, Liu, F. Extreme low dose of 5-fluorouracil reverses MDR in cancer by sensitizing cancer associated fibroblasts and down-regulating P-gp. PLoS One 2017;12:e0180023. https://doi.org/10.1371/journal.pone.0180023.Search in Google Scholar PubMed PubMed Central

102. Erin, N, Grahovac, J, Brozovic, A, Efferth, T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updates: Rev Comment Antimicrob Anticancer Chemother 2020;53:100715. https://doi.org/10.1016/j.drup.2020.100715.Search in Google Scholar PubMed

103. Naci, D, El Azreq, MA, Chetoui, N, Lauden, L, Sigaux, F, Charron, D, et al.. α2β1 integrin promotes chemoresistance against doxorubicin in cancer cells through extracellular signal-regulated kinase (ERK). J Biol Chem 2012;287:17065–76. https://doi.org/10.1074/jbc.m112.349365.Search in Google Scholar

104. Naik, A, Al-Yahyaee, A, Abdullah, N, Sam, JE, Al-Zeheimi, N, Yaish, MW, et al.. Neuropilin-1 promotes the oncogenic Tenascin-C/integrin β3 pathway and modulates chemoresistance in breast cancer cells. BMC Cancer 2018;18:533. https://doi.org/10.1186/s12885-018-4446-y.Search in Google Scholar PubMed PubMed Central

105. Jacobetz, MA, Chan, DS, Neesse, A, Bapiro, TE, Cook, N, Frese, KK, et al.. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013;62:112–20. https://doi.org/10.1136/gutjnl-2012-302529.Search in Google Scholar PubMed PubMed Central

106. Martin, J, Midgley, A, Meran, S, Woods, E, Bowen, T, Phillips, AO, et al.. Tumor necrosis factor-stimulated gene 6 (TSG-6)-mediated interactions with the inter-α-inhibitor heavy chain 5 facilitate tumor growth factor β1 (TGFβ1)-dependent fibroblast to myofibroblast differentiation. J Biol Chem 2016;291:13789–801. https://doi.org/10.1074/jbc.M115.670521.Search in Google Scholar PubMed PubMed Central

107. McCarthy, JB, El-Ashry, D, Turley, EA. Hyaluronan, cancer-associated fibroblasts and the tumor microenvironment in malignant progression. Front Cell Dev Biol 2018;6:48. https://doi.org/10.3389/fcell.2018.00048.Search in Google Scholar PubMed PubMed Central

108. Zhang, G, He, Y, Liu, Y, Du, Y, Yang, C, Gao, F. Reduced hyaluronan cross-linking induces breast cancer malignancy in a CAF-dependent manner. Cell Death Dis 2021;12:586. https://doi.org/10.1038/s41419-021-03875-6.Search in Google Scholar PubMed PubMed Central

109. Zhang, D, Li, L, Jiang, H, Knolhoff, BL, Lockhart, AC, Wang-Gillam, A, et al.. Constitutive IRAK4 activation underlies poor prognosis and chemoresistance in pancreatic ductal adenocarcinoma. Clin Cancer Res 2017;23:1748–59. https://doi.org/10.1158/1078-0432.ccr-16-1121.Search in Google Scholar

110. Hessmann, E, Patzak, MS, Klein, L, Chen, N, Kari, V, Ramu, I, et al.. Fibroblast drug scavenging increases intratumoural gemcitabine accumulation in murine pancreas cancer. Gut 2018;67:497–507. https://doi.org/10.1136/gutjnl-2016-311954.Search in Google Scholar PubMed PubMed Central

111. Zhang, D, Li, L, Jiang, H, Li, Q, Wang-Gillam, A, Yu, J, et al.. Tumor-stroma IL1β-IRAK4 feedforward circuitry drives tumor fibrosis, chemoresistance, and poor prognosis in pancreatic cancer. Cancer Res 2018;78:1700–12. https://doi.org/10.1158/0008-5472.can-17-1366.Search in Google Scholar PubMed PubMed Central

112. Mathot, P, Grandin, M, Devailly, G, Souaze, F, Cahais, V, Moran, S, et al.. DNA methylation signal has a major role in the response of human breast cancer cells to the microenvironment. Oncogenesis 2017;6:e390. https://doi.org/10.1038/oncsis.2017.88.Search in Google Scholar PubMed PubMed Central

113. Huang, WC, Yadav, VK, Cheng, WH, Wang, CH, Hsieh, MS, Huang, TY, et al.. The MEK/ERK/miR-21 signaling is critical in Osimertinib resistance in EGFR-mutant non-small cell lung cancer cells. Cancers 2021;13:6005. https://doi.org/10.3390/cancers13236005.Search in Google Scholar PubMed PubMed Central

114. Sirica, AE. The role of cancer-associated myofibroblasts in intrahepatic cholangiocarcinoma. Nat Rev Gastroenterol Hepatol 2011;9:44–54. https://doi.org/10.1038/nrgastro.2011.222.Search in Google Scholar PubMed

115. Chung, AS, Ferrara, N. Developmental and pathological angiogenesis. Annu Rev Cell Dev Biol 2011;27:563–84. https://doi.org/10.1146/annurev-cellbio-092910-154002.Search in Google Scholar PubMed

116. Park, JS, Kim, IK, Han, S, Park, I, Kim, C, Bae, J, et al.. Normalization of tumor vessels by Tie2 activation and Ang2 inhibition enhances drug delivery and produces a favorable tumor microenvironment. Cancer Cell 2017;31:157–8. https://doi.org/10.1016/j.ccell.2016.12.009.Search in Google Scholar PubMed

117. Tan, Q, Saggar, JK, Yu, M, Wang, M, Tannock, IF. Mechanisms of drug resistance related to the microenvironment of solid tumors and possible strategies to inhibit them. Cancer J 2015;21:254–62. https://doi.org/10.1097/ppo.0000000000000131.Search in Google Scholar PubMed

118. Wu, T, Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett 2017;387:61–8. https://doi.org/10.1016/j.canlet.2016.01.043.Search in Google Scholar PubMed

119. Zhang, L, Hannay, JA, Liu, J, Das, P, Zhan, M, Nguyen, T, et al.. Vascular endothelial growth factor overexpression by soft tissue sarcoma cells: implications for tumor growth, metastasis, and chemoresistance. Cancer Res 2006;66:8770–8. https://doi.org/10.1158/0008-5472.can-06-1217.Search in Google Scholar

120. Harris, AL. Angiogenesis as a new target for cancer control. EJC Suppl 2003;1:1–12. https://doi.org/10.1016/s1359-6349(03)00007-7.Search in Google Scholar

121. Folkman, J. Opinion - angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov 2007;6:273–86. https://doi.org/10.1038/nrd2115.Search in Google Scholar PubMed

122. Moya Garcia, AA, Morilla, I, Ranea, JAG. Oncogenic signalling networks and polypharmacology as paradigms to cope with cancer heterogeneity. Curr Proteonomics 2014;11:210–7. https://doi.org/10.2174/157016461103140922164228.Search in Google Scholar

123. Najafi, M, Farhood, B, Mortezaee, K. Cancer stem cells (CSCs) in cancer progression and therapy. J Cell Physiol 2019;234:8381–95. https://doi.org/10.1002/jcp.27740.Search in Google Scholar PubMed

124. Tang, YA, Chen, YF, Bao, Y, Mahara, S, Yatim, S, Oguz, G, et al.. Hypoxic tumor microenvironment activates GLI2 via HIF-1α and TGF-β2 to promote chemoresistance in colorectal cancer. Proc Natl Acad Sci USA 2018;115:E5990–e99. https://doi.org/10.1073/pnas.1801348115.Search in Google Scholar PubMed PubMed Central

125. Chen, Z, Shi, T, Zhang, L, Zhu, P, Deng, M, Huang, C, et al.. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: a review of the past decade. Cancer Lett 2016;370:153–64. https://doi.org/10.1016/j.canlet.2015.10.010.Search in Google Scholar PubMed

126. El-Awady, R, Saleh, E, Hashim, A, Soliman, N, Dallah, A, Elrasheed, A, et al.. The role of eukaryotic and prokaryotic ABC transporter family in failure of chemotherapy. Front Pharmacol 2016;7:535–5. https://doi.org/10.3389/fphar.2016.00535.Search in Google Scholar PubMed PubMed Central

127. Assaraf, YG, Brozovic, A, Goncalves, AC, Jurkovicova, D, Line, A, Machuqueiro, M, et al.. The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist Updates 2019;46:100645. https://doi.org/10.1016/j.drup.2019.100645.Search in Google Scholar PubMed

128. Dean, M, Fojo, T, Bates, S. Tumour stem cells and drug resistance. Nat Rev Cancer 2005;5:275–84. https://doi.org/10.1038/nrc1590.Search in Google Scholar PubMed

129. Steinbichler, TB, Dudás, J, Skvortsov, S, Ganswindt, U, Riechelmann, H, SkvortsovaII. Therapy resistance mediated by cancer stem cells. Semin Cancer Biol 2018;53:156–67. https://doi.org/10.1016/j.semcancer.2018.11.006.Search in Google Scholar PubMed

130. Dzobo, K, Senthebane, DA, Rowe, A, Thomford, NE, Mwapagha, LM, Al-Awwad, N, et al.. Cancer stem cell hypothesis for therapeutic innovation in clinical Oncology? Taking the root out, not chopping the leaf. OMICS A J Integr Biol 2016;20:681–91. https://doi.org/10.1089/omi.2016.0152.Search in Google Scholar PubMed

131. Shiotani, B, Kobayashi, M, Watanabe, M, Yamamoto, K, Sugimura, T, Wakabayashi, K. Involvement of the ATR- and ATM-dependent checkpoint responses in cell cycle arrest evoked by pierisin-1. Mol Cancer Res: MCR 2006;4:125–33. https://doi.org/10.1158/1541-7786.mcr-05-0104.Search in Google Scholar

132. Soeda, A, Park, M, Lee, D, Mintz, A, Androutsellis-Theotokis, A, McKay, RD, et al.. Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1alpha. Oncogene 2009;28:3949–59. https://doi.org/10.1038/onc.2009.252.Search in Google Scholar PubMed

133. Jiang, J, Feng, X, Zhou, W, Wu, Y, Yang, Y. MiR-128 reverses the gefitinib resistance of the lung cancer stem cells by inhibiting the c-met/PI3K/AKT pathway. Oncotarget 2016;7:73188–99. https://doi.org/10.18632/oncotarget.12283.Search in Google Scholar PubMed PubMed Central

134. Vermeulen, L, De Sousa, EMF, van der Heijden, M, Cameron, K, de Jong, JH, Borovski, T, et al.. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol 2010;12:468–76. https://doi.org/10.1038/ncb2048.Search in Google Scholar PubMed

135. Su, S, Chen, J, Yao, H, Liu, J, Yu, S, Lao, L, et al.. CD10(+)GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 2018;172:841–56.e16. https://doi.org/10.1016/j.cell.2018.01.009.Search in Google Scholar PubMed

136. Wu, Q, Tu, H, Li, J. Multifaceted roles of chemokine C-X-C motif ligand 7 in inflammatory diseases and cancer. Front Pharmacol 2022;13:914730. https://doi.org/10.3389/fphar.2022.914730.Search in Google Scholar PubMed PubMed Central

137. Beatty, GL, Gladney, WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res 2015;21:687–92. https://doi.org/10.1158/1078-0432.ccr-14-1860.Search in Google Scholar

138. Hu, Z. Chapter 11 – Using CAR-NK cells to overcome the host resistance to antibody immunotherapy and immune checkpoint blockade therapy. In: Bonavida, B, Jewett, A, editors. Successes and challenges of NK immunotherapy. Los Angeles: Academic Press; 2021:193–212 pp.10.1016/B978-0-12-824375-6.00010-2Search in Google Scholar

139. Jung, HI, Jeong, D, Ji, S, Ahn, TS, Bae, SH, Chin, S, et al.. Overexpression of PD-L1 and PD-L2 is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer research and treatment 2017;49:246–54. https://doi.org/10.4143/crt.2016.066.Search in Google Scholar PubMed PubMed Central

140. Pol, J, Vacchelli, E, Aranda, F, Castoldi, F, Eggermont, A, Cremer, I, et al.. Trial Watch: immunogenic cell death inducers for anticancer chemotherapy. OncoImmunology 2015;4:e1008866. https://doi.org/10.1080/2162402x.2015.1008866.Search in Google Scholar PubMed PubMed Central

141. Borghaei, H, Langer, CJ, Gadgeel, S, Papadimitrakopoulou, VA, Patnaik, A, Powell, SF, et al.. 24-Month overall survival from KEYNOTE-021 Cohort G: pemetrexed and carboplatin with or without pembrolizumab as first-line therapy for advanced nonsquamous non-small cell lung cancer. J Thorac Oncol 2019;14:124–9.10.1016/j.jtho.2018.08.004Search in Google Scholar PubMed

142. Gadgeel, S, Rodríguez-Abreu, D, Speranza, G, Esteban, E, Felip, E, Dómine, M, et al.. Updated analysis from KEYNOTE-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer. J Clin Oncol: Off J Am Soc Clin Oncol 2020;38:1505–17. https://doi.org/10.1200/jco.19.03136.Search in Google Scholar

143. Cortes, J, Cescon, DW, Rugo, HS, Nowecki, Z, Im, SA, Yusof, MM, et al.. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet (London, England) 2020;396:1817–28. https://doi.org/10.1016/s0140-6736(20)32531-9.Search in Google Scholar

144. Reck, M, Mok, TSK, Nishio, M, Jotte, RM, Cappuzzo, F, Orlandi, F, et al.. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med 2019;7:387–401. https://doi.org/10.1016/s2213-2600(19)30084-0.Search in Google Scholar PubMed

145. Brenner, AJ, Floyd, J, Fichtel, L, Michalek, J, Kanakia, KP, Huang, S, et al.. Phase 2 trial of hypoxia activated evofosfamide (TH302) for treatment of recurrent bevacizumab-refractory glioblastoma. Sci Rep 2021;11:2306. https://doi.org/10.1038/s41598-021-81841-0.Search in Google Scholar PubMed PubMed Central

146. Powles, T, Plimack, ER, Soulières, D, Waddell, T, Stus, V, Gafanov, R, et al.. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol 2020;21:1563–73. https://doi.org/10.1016/s1470-2045(20)30436-8.Search in Google Scholar PubMed

147. Choueiri, TK, Motzer, RJ, Rini, BI, Haanen, J, Campbell, MT, Venugopal, B, et al.. Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma. Ann Oncol 2020;31:1030–9. https://doi.org/10.1016/j.annonc.2020.04.010.Search in Google Scholar PubMed PubMed Central

148. Taylor, MH, Lee, CH, Makker, V, Rasco, D, Dutcus, CE, Wu, J, et al.. Phase IB/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol: Off J Am Soc Clin Oncol 2020;38:1154–63. https://doi.org/10.1200/jco.19.01598.Search in Google Scholar

149. Dillon, MT, Boylan, Z, Smith, D, Guevara, J, Mohammed, K, Peckitt, C, et al.. PATRIOT: a phase I study to assess the tolerability, safety and biological effects of a specific ataxia telangiectasia and Rad3-related (ATR) inhibitor (AZD6738) as a single agent and in combination with palliative radiation therapy in patients with solid tumours. Clin Transl Radiat Oncol 2018;12:16–20. https://doi.org/10.1016/j.ctro.2018.06.001.Search in Google Scholar PubMed PubMed Central

150. Toor, SM, Sasidharan Nair, V, Decock, J, Elkord, E. Immune checkpoints in the tumor microenvironment. Semin Cancer Biol 2020;65:1–12. https://doi.org/10.1016/j.semcancer.2019.06.021.Search in Google Scholar PubMed

151. Rolinski, J, Hus, I. Breaking immunotolerance of tumors: a new perspective for dendritic cell therapy. J Immunot 2014;11:311–8. https://doi.org/10.3109/1547691x.2013.865094.Search in Google Scholar

152. Reizis, B. Plasmacytoid dendritic cells: development, regulation, and function. Immunity 2019;50:37–50. https://doi.org/10.1016/j.immuni.2018.12.027.Search in Google Scholar PubMed PubMed Central

153. Sabree, SA, Voigt, AP, Blackwell, SE, Vishwakarma, A, Chimenti, MS, Salem, AK, et al.. Direct and indirect immune effects of CMP-001, a virus-like particle containing a TLR9 agonist. J Immunother Cancer 2021;9:e002484. https://doi.org/10.1136/jitc-2021-002484.Search in Google Scholar PubMed PubMed Central

154. Milhem, M, Zakharia, Y, Davar, D, Buchbinder, E, Medina, T, Daud, A, et al.. 304 Intratumoral injection of CMP-001, a toll-like receptor 9 (TLR9) agonist, in combination with pembrolizumab reversed programmed death receptor 1 (PD-1) blockade resistance in advanced melanoma. J Immunother Cancer 2020;8:A186–7.10.1136/jitc-2020-SITC2020.0304Search in Google Scholar

155. Romee, R, Cooley, S, Berrien-Elliott, MM, Westervelt, P, Verneris, MR, Wagner, JE, et al.. First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 2018;131:2515–27. https://doi.org/10.1182/blood-2017-12-823757.Search in Google Scholar PubMed PubMed Central

156. Wrangle, JM, Velcheti, V, Patel, MR, Garrett-Mayer, E, Hill, EG, Ravenel, JG, et al.. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol 2018;19:694–704. https://doi.org/10.1016/s1470-2045(18)30148-7.Search in Google Scholar PubMed PubMed Central

157. Diab, A, Tykodi, SS, Daniels, GA, Maio, M, Curti, BD, Lewis, KD, et al.. Bempegaldesleukin plus nivolumab in first-line metastatic melanoma. J Clin Oncol 2021;39:2914–25. https://doi.org/10.1200/jco.21.00675.Search in Google Scholar PubMed PubMed Central

158. Mayor, P, Starbuck, K, Zsiros, E. Adoptive cell transfer using autologous tumor infiltrating lymphocytes in gynecologic malignancies. Gynecol Oncol 2018;150:361–9. https://doi.org/10.1016/j.ygyno.2018.05.024.Search in Google Scholar PubMed PubMed Central

159. Jazaeri, AA, Zsiros, E, Amaria, RN, Artz, AS, Edwards, RP, Wenham, RM, et al.. Safety and efficacy of adoptive cell transfer using autologous tumor infiltrating lymphocytes (LN-145) for treatment of recurrent, metastatic, or persistent cervical carcinoma. J Clin Oncol 2019;37:2538. https://doi.org/10.1200/jco.2019.37.15_suppl.2538.Search in Google Scholar

160. Sarnaik, AA, Hamid, O, Khushalani, NI, Lewis, KD, Medina, T, Kluger, HM, et al.. Lifileucel, a tumor-infiltrating lymphocyte therapy, in metastatic melanoma. J Clin Oncol: Off J Am Soc Clin Oncol 2021;39:2656–66. https://doi.org/10.1200/jco.21.00612.Search in Google Scholar

161. Zhang, C, Zhou, X, Zhang, H, Han, X, Li, B, Yang, R, et al.. Recent progress of novel nanotechnology challenging the multidrug resistance of cancer. Front Pharmacol 2022;13:776895. https://doi.org/10.3389/fphar.2022.776895.Search in Google Scholar PubMed PubMed Central

162. Yang, S, Gao, H. Nanoparticles for modulating tumor microenvironment to improve drug delivery and tumor therapy. Pharmacol Res 2017;126:97–108. https://doi.org/10.1016/j.phrs.2017.05.004.Search in Google Scholar PubMed

163. You, Q, Zhang, K, Liu, J, Liu, C, Wang, H, Wang, M, et al.. Persistent regulation of tumor hypoxia microenvironment via a bioinspired Pt-based oxygen nanogenerator for multimodal imaging-guided synergistic phototherapy. Adv Sci 2020;7:1903341. https://doi.org/10.1002/advs.201903341.Search in Google Scholar PubMed PubMed Central

164. Neophytou, CM, Panagi, M, Stylianopoulos, T, Papageorgis, P. The role of tumor microenvironment in cancer metastasis: molecular mechanisms and therapeutic opportunities. Cancers 2021;13:2053. https://doi.org/10.3390/cancers13092053.Search in Google Scholar PubMed PubMed Central

165. Yang, M, Li, J, Gu, P, Fan, X. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater 2021;6:1973–87. https://doi.org/10.1016/j.bioactmat.2020.12.010.Search in Google Scholar PubMed PubMed Central

166. Han, X, Wei, Q, Lv, Y, Weng, L, Huang, H, Wei, Q, et al.. Ginseng-derived nanoparticles potentiate immune checkpoint antibody efficacy by reprogramming the cold tumor microenvironment. Mol Ther 2022;30:327–40. https://doi.org/10.1016/j.ymthe.2021.08.028.Search in Google Scholar PubMed PubMed Central

167. Sun, D, Zou, Y, Song, L, Han, S, Yang, H, Chu, D, et al.. A cyclodextrin-based nanoformulation achieves co-delivery of ginsenoside Rg3 and quercetin for chemo-immunotherapy in colorectal cancer. Acta Pharm Sin B 2022;12:378–93. https://doi.org/10.1016/j.apsb.2021.06.005.Search in Google Scholar PubMed PubMed Central

168. Wang, S, Yang, Y, Wu, H, Li, J, Xie, P, Xu, F, et al.. Thermosensitive and tum or microenvironment activated nanotheranostics for the chemodynamic/photothermal therapy of colorectal tumor. J Colloid Interface Sci 2022;612:223–34. https://doi.org/10.1016/j.jcis.2021.12.126.Search in Google Scholar PubMed

169. Xiao, Y, Chen, J, Zhou, H, Zeng, X, Ruan, Z, Pu, Z, et al.. Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy. Nat Commun 2022;13:758.10.1038/s41467-022-28279-8Search in Google Scholar PubMed PubMed Central

170. Wang, Q, Shao, X, Zhang, Y, Zhu, M, Wang, FXC, Mu, J, et al.. Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med 2023;12:11149–65. https://doi.org/10.1002/cam4.5698.Search in Google Scholar PubMed PubMed Central

171. Zhen, W, Weichselbaum, RR, Lin, W. Nanoparticle-mediated radiotherapy remodels the tumor microenvironment to enhance antitumor efficacy. Adv Mater 2023;35:e2206370. https://doi.org/10.1002/adma.202206370.Search in Google Scholar PubMed PubMed Central

172. An, J, Liu, M, Zhao, L, Lu, W, Wu, S, Zhang, K, et al.. Boosting tumor immunotherapy by bioactive nanoparticles via Ca2+ interference mediated TME reprogramming and specific PD-L1 depletion. Adv Funct Mater 2022;32:2201275. https://doi.org/10.1002/adfm.202201275.Search in Google Scholar

173. Ni, R, Huang, L, Li, Z, Zhang, W, Wang, Y, Shen, Y, et al.. Multifunctional ROS-responsive and TME-modulated lipid-polymer hybrid nanoparticles for enhanced tumor penetration. Int J Nanomed 2022;17:5883–97. https://doi.org/10.2147/ijn.s383517.Search in Google Scholar

174. Ramzy, A, Soliman, AH, Hassanein, SI, Sebak, AA. Multitarget, multiagent PLGA nanoparticles for simultaneous tumor eradication and TME remodeling in a melanoma mouse model. Drug Deliv Transl Res 2023. https://doi.org/10.1007/s13346-023-01413-9. 37612575Search in Google Scholar PubMed PubMed Central

175. Kalave, S, Hegde, N, Juvale, K. Applications of nanotechnology-based approaches to overcome multi-drug resistance in cancer. Curr Pharmaceut Des 2022;28:3140–57. https://doi.org/10.2174/1381612828666220401142300.Search in Google Scholar PubMed

176. Rivankar, S. An overview of doxorubicin formulations in cancer therapy. J Cancer Res Therapeut 2014;10:853–8. https://doi.org/10.4103/0973-1482.139267.Search in Google Scholar PubMed

177. Gianni, L, Mansutti, M, Anton, A, Calvo, L, Bisagni, G, Bermejo, B, et al.. Comparing neoadjuvant nab-paclitaxel vs paclitaxel both followed by anthracycline regimens in women with ERBB2/HER2-negative breast cancer-the evaluating treatment with neoadjuvant Abraxane (ETNA) trial: a randomized phase 3 clinical trial. JAMA Oncol 2018;4:302–8. https://doi.org/10.1001/jamaoncol.2017.4612.Search in Google Scholar PubMed PubMed Central

178. Tian, X, Nguyen, M, Foote, HP, Caster, JM, Roche, KC, Peters, CG, et al.. CRLX101, a nanoparticle-drug conjugate containing camptothecin, improves rectal cancer chemoradiotherapy by inhibiting DNA repair and HIF1α. Cancer Res 2017;77:112–22. https://doi.org/10.1158/0008-5472.can-15-2951.Search in Google Scholar

179. Ma, WW, Tolcher, AW, Strauss, JF, Bekaii-Saab, TS, Zhao, Y, Perez, CA, et al.. ELU-FRα-1: a study to evaluate ELU001 in patients with solid tumors that overexpress folate receptor alpha (FRα). J Clin Oncol 2022;40:TPS3158–8. https://doi.org/10.1200/jco.2022.40.16_suppl.tps3158.Search in Google Scholar

180. Chen, X, Zhou, J, Li, X, Wang, X, Lin, Y, Wang, X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett 2018;435:80–91. https://doi.org/10.1016/j.canlet.2018.08.001.Search in Google Scholar PubMed

181. Xu, R, Rai, A, Chen, M, Suwakulsiri, W, Greening, DW, Simpson, RJ. Extracellular vesicles in cancer - implications for future improvements in cancer care. Nat Rev Clin Oncol 2018;15:617–38. https://doi.org/10.1038/s41571-018-0036-9.Search in Google Scholar PubMed

182. Roma-Rodrigues, C, Mendes, R, Baptista, PV, Fernandes, AR. Targeting tumor microenvironment for cancer therapy. Int J Mol Sci 2019;20:840. https://doi.org/10.3390/ijms20040840.Search in Google Scholar PubMed PubMed Central

183. Deepak, KGK, Vempati, R, Nagaraju, GP, Dasari, VR, Nagini, S, Rao, DN, et al.. Tumor microenvironment: challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol Res 2020;153:104683. https://doi.org/10.1016/j.phrs.2020.104683.Search in Google Scholar PubMed

184. Salemme, V, Centonze, G, Cavallo, F, Defilippi, P, Conti, L. The crosstalk between tumor cells and the immune microenvironment in breast cancer: implications for immunotherapy. Front Oncol 2021;11:11. https://doi.org/10.3389/fonc.2021.610303.Search in Google Scholar PubMed PubMed Central

185. Li, J, Zhang, G, Liu, C-G, Xiang, X, Le, MTN, Sethi, G, et al.. The potential role of exosomal circRNAs in the tumor microenvironment: insights into cancer diagnosis and therapy. Theranostics 2022;12:87–104. https://doi.org/10.7150/thno.64096.Search in Google Scholar PubMed PubMed Central

186. Tang, Q, Yang, S, He, G, Zheng, H, Zhang, S, Liu, J, et al.. Tumor-derived exosomes in the cancer immune microenvironment and cancer immunotherapy. Cancer Lett 2022;548:215823.10.1016/j.canlet.2022.215823Search in Google Scholar PubMed

187. Thakur, A, Johnson, A, Jacobs, E, Zhang, K, Chen, J, Wei, Z, et al.. Energy sources for exosome communication in a cancer microenvironment. Cancers 2022;14:1698. https://doi.org/10.3390/cancers14071698.Search in Google Scholar PubMed PubMed Central

188. Baronzio, G, Parmar, G, Baronzio, M. Overview of methods for overcoming hindrance to drug delivery to tumors, with special attention to tumor interstitial fluid. Front Oncol 2015;5:5. https://doi.org/10.3389/fonc.2015.00165.Search in Google Scholar PubMed PubMed Central

189. Huang, YL, Segall, JE, Wu, M. Microfluidic modeling of the biophysical microenvironment in tumor cell invasion. Lab Chip 2017;17:3221–33. https://doi.org/10.1039/c7lc00623c.Search in Google Scholar PubMed PubMed Central

190. Wang, M, Zhao, J, Zhang, L, Wei, F, Lian, Y, Wu, Y, et al.. Role of tumor microenvironment in tumorigenesis. J Cancer 2017;8:761–73. https://doi.org/10.7150/jca.17648.Search in Google Scholar PubMed PubMed Central

191. Clegg, J, Koch, MK, Thompson, EW, Haupt, LM, Kalita-de Croft, P, Bray, LJ. Three-dimensional models as a new frontier for studying the role of proteoglycans in the normal and malignant breast microenvironment. Front Cell Dev Biol 2020;8:8. https://doi.org/10.3389/fcell.2020.569454.Search in Google Scholar PubMed PubMed Central

192. Feng, Y, Liao, Z, Zhang, H, Xie, X, You, F, Liao, X, et al.. Emerging nanomedicines strategies focused on tumor microenvironment against cancer recurrence and metastasis. Chem Eng J 2023;452:139506. https://doi.org/10.1016/j.cej.2022.139506.Search in Google Scholar

193. Salgaller, ML. Technology evaluation: Bevacizumab, Genentech/Roche. Curr Opin Mol Therapeut 2003;5:657–67.Search in Google Scholar

194. Song, Y, Fu, Y, Xie, Q, Zhu, B, Wang, J, Zhang, B. Anti-angiogenic agents in combination with immune checkpoint inhibitors: a promising strategy for cancer treatment. Front Immunol 2020;11:1956. https://doi.org/10.3389/fimmu.2020.01956.Search in Google Scholar PubMed PubMed Central

195. Chauhan, VP, Stylianopoulos, T, Martin, JD, Popović, Z, Chen, O, Kamoun, WS, et al.. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat Nanotechnol 2012;7:383–8. https://doi.org/10.1038/nnano.2012.45.Search in Google Scholar PubMed PubMed Central

196. Sakurai, Y, Hada, T, Yamamoto, S, Kato, A, Mizumura, W, Harashima, H. Remodeling of the extracellular matrix by endothelial cell-targeting siRNA improves the EPR-based delivery of 100 nm particles. Mol Ther 2016;24:2090–9. https://doi.org/10.1038/mt.2016.178.Search in Google Scholar PubMed PubMed Central

197. Markovic, SN, Suman, VJ, Javed, A, Reid, JM, Wall, DJ, Erickson, LA, et al.. Sequencing ipilimumab immunotherapy before or after chemotherapy (Nab-Paclitaxel and Bevacizumab) for the treatment of BRAFwt (BRAF wild-type) metastatic malignant melanoma: results of a study of academic and community cancer research united (ACCRU) RU261206I. Am J Clin Oncol 2020;43:115–21. https://doi.org/10.1097/coc.0000000000000644.Search in Google Scholar PubMed

198. Diop-Frimpong, B, Chauhan, VP, Krane, S, Boucher, Y, Jain, RK. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci USA 2011;108:2909–14. https://doi.org/10.1073/pnas.1018892108.Search in Google Scholar PubMed PubMed Central

199. Stylianopoulos, T, Munn, LL, Jain, RK. Reengineering the tumor vasculature: improving drug delivery and efficacy. Trends in cancer 2018;4:258–9. https://doi.org/10.1016/j.trecan.2018.02.010.Search in Google Scholar PubMed PubMed Central

200. Zhao, Y, Cao, J, Melamed, A, Worley, M, Gockley, A, Jones, D, et al.. Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma. Proc Natl Acad Sci USA 2019;116:2210–9. https://doi.org/10.1073/pnas.1818357116.Search in Google Scholar PubMed PubMed Central

201. Li, W, Li, S, Chen, IX, Liu, Y, Ramjiawan, RR, Leung, CH, et al.. Combining losartan with radiotherapy increases tumor control and inhibits lung metastases from a HER2/neu-positive orthotopic breast cancer model. Radiat Oncol 2021;16:48. https://doi.org/10.1186/s13014-021-01775-9.Search in Google Scholar PubMed PubMed Central

202. Murphy, JE, Wo, JY, Ryan, DP, Clark, JW, Jiang, W, Yeap, BY, et al.. Total neoadjuvant therapy with fOLFIRINOX in combination with Losartan followed by chemoradiotherapy for locally advanced pancreatic cancer: a phase 2 clinical trial. JAMA Oncol 2019;5:1020–7. https://doi.org/10.1001/jamaoncol.2019.0892.Search in Google Scholar PubMed PubMed Central

203. Martin, JD, Panagi, M, Wang, C, Khan, TT, Martin, MR, Voutouri, C, et al.. Dexamethasone increases cisplatin-loaded nanocarrier delivery and efficacy in metastatic breast cancer by normalizing the tumor microenvironment. ACS Nano 2019;13:6396–408. https://doi.org/10.1021/acsnano.8b07865.Search in Google Scholar PubMed

204. Masterson, R, Hewitson, TD, Kelynack, K, Martic, M, Parry, L, Bathgate, R, et al.. Relaxin down-regulates renal fibroblast function and promotes matrix remodelling in vitro. Nephrol Dial Transplant 2004;19:544–52. https://doi.org/10.1093/ndt/gfg598.Search in Google Scholar PubMed

205. Kim, JH, Shin, BC, Park, WS, Lee, J, Kuh, H-J. Antifibrotic effects of pentoxifylline improve the efficacy of gemcitabine in human pancreatic tumor xenografts. Cancer Sci 2017;108:2470–7. https://doi.org/10.1111/cas.13405.Search in Google Scholar PubMed PubMed Central

206. Scott, AM, Wiseman, G, Welt, S, Adjei, A, Lee, FT, Hopkins, W, et al.. A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin Cancer Res 2003;9:1639–47.Search in Google Scholar

207. Jiang, H, Hegde, S, Knolhoff, BL, Zhu, Y, Herndon, JM, Meyer, MA, et al.. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med 2016;22:851–60. https://doi.org/10.1038/nm.4123.Search in Google Scholar PubMed PubMed Central

208. Waldhauer, I, Gonzalez-Nicolini, V, Freimoser-Grundschober, A, Nayak, TK, Fahrni, L, Hosse, RJ, et al.. Simlukafusp alfa (FAP-IL2v) immunocytokine is a versatile combination partner for cancer immunotherapy. mAbs 2021;13:1913791. https://doi.org/10.1080/19420862.2021.1913791.Search in Google Scholar PubMed PubMed Central


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/oncologie-2023-0459).


Received: 2023-10-23
Accepted: 2023-12-07
Published Online: 2024-01-03

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Frontmatter
  2. Review Articles
  3. Advances in ferroptosis of cancer therapy
  4. Immunotherapy in hepatocellular carcinoma: an overview of immune checkpoint inhibitors, drug resistance, and adverse effects
  5. The role of matrix metalloproteinase-2 in the metastatic cascade: a review
  6. The tumor microenvironment: a key player in multidrug resistance in cancer
  7. Robotic vs. laparoscopic approach in obese patients with endometrial cancer: which is the best? A mini-review
  8. SLC25 family with energy metabolism and immunity in malignant tumors
  9. Research Articles
  10. Catalase expression is an independent prognostic marker in liver hepatocellular carcinoma
  11. A novel immune-associated prognostic signature based on the immune cell infiltration analysis for hepatocellular carcinoma
  12. AKAP12 inhibits the proliferation of ovarian cancer by activating the Hippo pathway
  13. AQP1 as a novel biomarker to predict prognosis and tumor immunity in glioma patients
  14. Exosomal circular RNA NT5E driven by heterogeneous nuclear ribonucleoprotein A1 induces temozolomide resistance by targeting microRNA-153 in glioma cells
  15. miR‐30a‐3p inhibits the proliferation of laryngeal cancer cells by targeting DNMT3a through regulating DNA methylation of PTEN
  16. Disulfidptosis-related long non-coding RNAs predict prognosis and indicate therapeutic response in non-small cell lung carcinoma
  17. Case Report
  18. Primary retroperitoneal choriocarcinoma with lung and liver metastasis in a male patient: case report
  19. Short Commentary
  20. Clinical pharmacy services in cancer patients with hypertension
Downloaded on 13.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2023-0459/html
Scroll to top button