Tumor microenvironment

Last updated
Component of the tumor microenvironment (TME). The tumor microenvironment is a complex system of various tumor cells, stromal cells, and immune cells. Components-of-the-tumor-microenvironment.png
Component of the tumor microenvironment (TME). The tumor microenvironment is a complex system of various tumor cells, stromal cells, and immune cells.

The tumor microenvironment is a complex ecosystem surrounding a tumor, composed of cancer cells, stromal tissue (including blood vessels, immune cells, fibroblasts and signaling molecules) and the extracellular matrix. [2] [3] [4] [5] Mutual interaction between cancer cells and the different components of the tumor microenvironment support its growth and invasion in healthy tissues which correlates with tumor resistance to current treatments and poor prognosis. The tumor microenvironment is in constant change because of the tumor's ability to influence the microenvironment by releasing extracellular signals, promoting tumor angiogenesis and inducing peripheral immune tolerance, while the immune cells in the microenvironment can affect the growth and evolution of cancerous cells. [2] [6] [7] [8] [9]

Contents

History

The concept of the tumor microenvironment (TME) dates back to 1863 when Rudolf Virchow established a connection between inflammation and cancer. However, it wasn't until 1889 that Stephen Paget's "seed and soil" theory introduced the important role of TME in cancer metastasis, highlighting the intricate relationship between tumors and their surrounding microenvironment. The theory indicated that cancer cells have tendencies when spreading. Paget proposed that the metastases of a particular type of cancer ("the seed") often metastasizes to certain sites ("the soil") based on the similarity of the original and secondary tumor sites. In other words, just as seeds need fertile soil to grow, cancer cells require a supportive microenvironment to metastasize. [10] [11]

In 1928, James Ewing challenged Paget's theory with his own perspective on cancer metastasis. Ewing proposed that the ability of cancer cells to metastasize was primarily influenced by mechanical mechanisms such as anatomical and hemodynamic factors of the vascular connection, with tumor cells more likely to be trapped in the first connected organ. [10] [12] This viewpoint suggested that certain properties or mutations within cancer cells might dictate their metastatic potential, independent of the surrounding tissue environment. [10] Isaiah Fidler formulated a complementary hypothesis in the 1970s, where he proposed that while the mechanical aspects of blood flow is important, metastatic colonization specifically targets certain organs, known as organotropism. [13]

In the late 1970s, attention shifted towards understanding the role of lymphocytes within the tumor microenvironment. Reports emerged detailing the presence and activities of tumor-infiltrating T and B lymphocytes, as well as natural killer (NK) cells. Researchers observed that tumor-infiltrating T cells had both anti-tumor cytotoxicity and immune-suppressive properties. However, their cytotoxic activity was found to be lower compared to lymphocytes from distant sites, likely due to the overall immunosuppressive state in tumor-bearing individuals. [14]

Vasculature

A tumor's vasculature is important to its growth, as blood vessels deliver oxygen, nutrients, and growth factors to the tumor. [15] Tumors smaller than 1–2 mm in diameter are delivered oxygen and nutrients through passive diffusion. In larger tumors the center becomes too far away from the existing blood supply, leading the tumor microenvironment to become hypoxic and acidic. [16] Angiogenesis is upregulated to feed the cancer cells and is linked to tumor malignancy. [17]

Endothelial cells and angiogenesis

In hypoxic environments the tissue sends out signals called hypoxia inducible factors (HIFs) that can stimulate nearby endothelial cells to secrete factors such as vascular endothelial growth factor (VEGF). VEGF activates the endothelial cells, which begins the process of angiogenesis, where new blood vessels emerge from pre-existing vasculature. [18] The blood vessel formed in the tumor environment often doesn't mature properly, and as a result the vasculature formed in the tumor microenvironment differs from that of normal tissue. The blood vessels formed are often "leaky" and tortuous, with a compromised blood flow. [19] [16] As tumors cannot grow large without proper vasculature, sustained angiogenesis is therefore considered one of the hallmarks of cancer. [20]

In later stages of tumor progression endothelial cells can differentiate into carcinoma associated fibroblasts, which furthers metastasis. [16]

Enhanced permeability and retention effect

The enhanced permeability and retention effect is the observation that the vasculature of tumors tend to accumulate macromolecules in the blood stream to a greater extent than in normal tissue. This is due to the "leaky" nature of the vasculature around tumors, and a lacking lymphatic system. [21] The permeable vasculature allows for easier delivery of therapeutic drugs to the tumor, and the lacking lymphatic vessels contribute to an increased retention. The permeable vasculature is thought to have several causes, including insufficient pericytes and a malformed basement membrane. [22]

Hypoxia

Tumor stroma and extracellular matrix in hypoxia Tumour stroma and extracellular matrix in hypoxia.svg
Tumor stroma and extracellular matrix in hypoxia

While angiogenesis can reduce the hypoxia in the tumor microenvironment, the partial pressure of oxygen is below 5 mmHg in over 50% of locally advanced solid tumors, compared to venous blood which has a partial pressure of oxygen at 40-60 mmHg. [18] [5] A hypoxic environment leads to genetic instability by downregulating genes involved in DNA repair mechanisms such as nucleotide excision repair and mismatch repair pathways. [23] This genetic instability leads to a high number of mutated cells, and is associated with cancer progression. [5] Periods of mild and acute hypoxia and reoxygenation can lead cancer cells to adapt and grow into more aggressive phenotypes. [18]

Hypoxia causes the upregulation of hypoxia induced factors (HIFs), which are transcription factors that decides how cells respond to a lack of oxygen. [16] HIFs induces the transcription of thousands of genes, some of which induces angiogenesis or furthers metastasis, leading, for instance, to increased cell migration and matrix remodeling. [24] [4] An increased HIF expression can lead tumor cells to shift their metabolism from aerobic to anaerobic, where they obtain energy through glycolysis. [25] Cells with an elevated glucose metabolism produce lactate, which decreases the pH in the microenvironment from a neutral and healthy 7.35-7.45 to an acidic 6.3-7.0. This phenomenon is described as the "Warburg effect". [25] [26] HIFs also regulate immune cells, and an increased expression can lead to the inactivation of anti-tumor functions. This furthers the survival of tumor cells and hinders anti-tumor treatment. [25]

Stromal cells

Cancer is a complex disease involving both tumor cells and surrounding stromal cells. In cancer biology, the stroma is defined as the nonmalignant cells found in the supportive tissue surrounding tumors. These cells include fibroblasts, immune cells, endothelial cells, and various other cell types. [27]

Stromal cells within the tumor microenvironment represent an important cellular component in cancer development, influencing tumor metabolism, growth, metastasis, immune evasion, and resistance to chemotherapy. These cells can originate from neighboring non-cancerous stromal cells or undergo transdifferentiation from tumor cells. Stromal cells contribute to tumor initiation, progression and drug resistance, and the stroma is known to evolve as the tumor develops. Understanding the interactions between cancer cells and stromal cells is essential for developing effective cancer treatments. [27] [28] Alterations in the stroma, including the activation of fibroblasts into carcinoma-associated fibroblasts (CAFs) and remodeling of the extracellular matrix (ECM), are recognized as important in cancer progression and potential targets for therapy and diagnosis. [29]

Carcinoma associated fibroblasts

Stromal cell types in early and late-stage tumors. Stromal cell in tumor microenvironment.jpg
Stromal cell types in early and late-stage tumors.

Carcinoma associated fibroblasts (CAFs) are a heterogenous group of activated fibroblasts central to the reactive stroma within the tumor microenvironment. The precise definition of CAFs remains challenging due to variations in cellular origins and expression markers. However, evidence suggests CAFs originate from activated resident fibroblasts, bone marrow-derived mesenchymal stem cells (MSCs), cancer cells undergoing epithelial-mesenchymal transition (ETM), or endothelial cells trough endothelial to mesenchymal transition (EndMT). [31] [32] [11]

CAFs are one of the most common components of the tumor stroma and are particularly found in the interstitial spaces of breast, prostate, and pancreatic cancer. [28] They interact with cancer cells by secreting a variety of extracellular matrix components or cell-cell adhesion, which is important in regulating the biological behavior of tumors. These regulations are particularly important for tumor development and influence cancer cell growth, invasion, inflammation, and angiogenesis. CAFs may also exhibit tumor-inhibitory properties in some cases. [28] [33] [11]

CAFs play a dual role in tumorigenesis; one that promotes tumor growth and another that inhibits it, with the former being more common and contributing to tumor development and therapy resistance through various mechanisms. [11] [33] Various subpopulations of CAFs have been identified across different cancer types. In breast cancer, for example, studies using single-cell RNA sequencing have revealed distinct phenotypes, including vascular CAFs, matrix CAFs, cycling CAFs, and developmental CAFs. [34] Studies using proteomic analysis and single-cell RNA sequencing have shed more light on the diverse characteristics of CAFs, revealing distinct and sometimes contradictory functions. Their functions appear to be context dependent. [31] This diversity in stomal composition not only shapes the tumor microenvironment, but also affects the behavior of tumor cells. [34] [35]

Targeting CAF has emerged as a promising strategy for improving cancer treatment, but the research faces several challenges. These include gaps in our understanding of CAF origins and their diverse functions, some of which may be helpful in combating tumors. [11] [33] [28]

Extracellular matrix remodeling

HIF regulates cancer cells HIF regulates interactions of cancer cells with ECM and ECM biosynthesis.svg
HIF regulates cancer cells

The extracellular matrix (ECM) is a tree-dimensional network of proteins and proteoglycans in the microenvironment and is present in all tissue. The ECM is a highly dynamic structure and is essential for tissue development, repair, support, and homeostasis. [36] [37] In healthy skin, the EMC is composed of various molecules such as collagens, glycoproteins, and glycosaminoglycans that regulate functions and mechanical properties. However, in tumors, the ECM plays an important role in shaping the tumor microenvironment and influences cancer progression, metastasis, and therapeutic response. This process is called extracellular matrix remodeling and is characterized by changes in protein content and enzymatic activity which influences signal transduction and cell-matrix alterations. [38] ECM remodeling involves dynamic alterations in ECM composition, organization, and biomechanical properties. ECM remodeling is induced by factors such as hypoxia, acidosis, inflammatory cells, or proteases secreted by tumor or stromal cells. [39]

Cellular mechanisms

Cells interact with and bind to the ECM through transmembrane receptors like integrins, discoidin domain receptor 2 (DDRs), and syndecans. The transmission of signals from the ECM to the cell interior involves various pathways. One primary way is direct transduction mediated by transmembrane proteins like integrins. [40] Integrins is the most studied ECM binding receptor and mediate ECM remodeling and regular cellular processes like proliferation, survival, migration, and invasion in response to ECM changes. They act as mechanotransducers by converting mechanical forces from the ECM or the cytoskeleton into chemical signals. Integrins can sense differences between simple, rigid two-dimensional surfaces and complex, malleable three-dimensional environments, altering cellular signaling accordingly. [41] [42]

In addition to integrins, other cell receptors like cell surface glycoprotein receptor (CD44), DDR2 and elastin-binding protein receptor (EBPR) can activate signaling pathways such as phosphatidylinositol 3-kinase and Akt. These receptors interact with various ECM components and create diverse cellular processes that contribute both to normal physiological functions and pathological conditions like cancer. [40]

Impact on cancer progression

Multiple factors determine whether tumor cells will be eliminated by the immune system or will escape detection. Tumor microenvironment.jpg
Multiple factors determine whether tumor cells will be eliminated by the immune system or will escape detection.

While ECM remodeling is tightly regulated under normal physiological conditions, it also modulates many of the tumor cell behaviors associated with cancer progression. This includes evasion of apoptosis, sustained angiogenesis, limitless replication potential, and tissue invasion. [43] [44] In cancer, changes in the ECM dynamics lead to changes in composition, density, and mechanical properties, affecting tumor aggressiveness and response to therapy. Research suggests that both pro- and anti-tumorigenic effects occurs during ECM remodeling. In early tumor formation, stromal cells produce excess ECM proteins, causing the tissue around the tumor to stiffen. Some of the contributing factors to tumor stiffness is increased type 1 collagen and acid deposition. [43] [45] Additionally, the restructured ECM and its degradation fragments (matrikines) impacts signaling pathways via cell-surface receptor interactions, leading to dysregulated stromal cell behavior and the emergence of an oncogenic microenvironment. [46]

Immune cells

Tumor-associated immune cells can be tumor-antagonizing or tumor-promoting, meaning that they can suppress or promote tumor growth. [47] Because of the effects of hypoxia, the anti-tumor abilities of many tumor-antagonizing immune cells, such as cytotoxic T cells and natural killer cells, become inhibited. Tumor-promoting immune cells such as regulatory T cells and myeloid derived suppressor cells will, on the other hand, become upregulated. [25]

Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models.svg
Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models
Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models.svg
Tumor-associated immune cells in the tumor microenvironment (TME) of breast cancer models
Immune checkpoints of immunosuppressive actions associated with breast cancer Immune checkpoints of immunosuppressive actions associated with breast cancer.svg
Immune checkpoints of immunosuppressive actions associated with breast cancer

Myeloid-derived suppressor cells and tumor-associated macrophages

Myeloid-derived suppressor cells are a heterogeneous population of cells of myelogenous origin [48] that are considered tumor promoting. [47] They have the potential to repress T cell responses, [49] can support angiogenesis by producing proteins such as vascular endothelial growth factor (VEGF), and can promote metastasis. Tumor associated macrophages with the M2 phenotype are considered myeloid-derived suppressor cells. [47]

Tumor-associated macrophages are a central component in the strong link between chronic inflammation and cancer, and are recruited to the tumor as a response to cancer-associated inflammation. [50] Their sluggish NF-κB activation allows for the smoldering inflammation seen in cancer. [51] Unlike normal macrophages, tumor-associated macrophages lack cytotoxic activity. [52] Monocyte derived macrophages are divided into inflammatory M1-polarized macrophages and anti-inflammatory M2-polarized macrophages. M1-polarized macrophages phagocytize tumor cells and are considered tumor-antagonizing. [16] M2-polarized macrophages are, on the other hand, tumor-promoting, because they promote tumor progression by suppressing immunosurveillance, [47] aiding angiogenesis by secreting vascular endothelial growth factor (VEGF) [5] and remodeling the extracellular matrix. [47] The tumor microenvironment promotes the M2-polarized macrophages, and an increased amount of tumor-associated macrophages is associated with worse prognosis. [16] [53] [54]

Tumor-associated macrophages are associated with using exosomes to deliver invasion-potentiating microRNA into cancerous cells, specifically breast cancer cells. [49] [55]

Neutrophils

Neutrophils are polymorphonuclear immune cells that are critical components of the innate immune system. Neutrophils can accumulate in tumors and in some cancers, such as lung adenocarcinoma, their abundance at the tumor site is associated with worsened disease prognosis. [56] [57] [58] Neutrophil numbers (and myeloid cell precursors) in the blood can be increased in some patients with solid tumors. [59] [60] [61]

Experiments in mice have mainly shown that tumor-associated neutrophils exhibit tumor-promoting functions, [62] [63] [64] [65] but a smaller number of studies show that neutrophils can also inhibit tumor growth. [66] [67] Tumor associated neutrophils can be divided into N1- and N2-polarized neutrophils. [47] N1-polarized neutrophils accumulate in the tumor in its early stages and support with tumor cell death. In later stages N2-polarized neutrophils promotes angiogenesis by secreting vascular endothelial growth factor (VEGF). [16]

Tumor infiltrating lymphocytes

Tumor infiltrating lymphocytes are lymphocytes, including T cells, B cells and natural killer cells, that penetrate the tumor and has the ability to recognize and kill cancer cells. [68] A high concentration is generally positively correlated with good prognosis (802). [69] This type of immune cells can also block metastasis, as natural killer cells are most efficient at killing cancer cells outside of the tumor microenvironment. [16] Tumor infiltrating lymphocytes has been used in therapeutic treatments, where lab amplificated immune cells are transferred to cancer patients to help their immune system fight the cancer. [69] This treatment has seen success in solid tumors such as melanoma. [70]

Tumor infiltrating lymphocytes can become tumor-promoting due to the immunosuppressive mechanisms of the tumor microenvironment. [69] Cancer cells induce apoptosis of activated T cells by secreting exosomes containing death ligands such as FasL and TRAIL, and via the same method, turn off the normal cytotoxic response of natural killer cells. [71] [72]

T cells

There are several types of T cells that are important to tumorigenesis, including cytotoxic T cells (CD8+), T helper 1 (Th-1) cells and regulatory T cells (Tregs). [16] CD8+ cells are tumor-antagonizing cells that recognize tumor antigens and targets cancer cells for destruction. In addition, CD8+ cells slow tumor progression and suppress angiogenesis by releasing interferon-gamma (IFN-γ). [16] Th-1 cells supports the activation and proliferation of CD8+ cells by secreting IFN-γ and interleukin-2 (IL-2), and by cross-presenting tumor antigens. [47] Tregs are, as opposed to CD8+, tumor promoting. They secrete tumor growth factors, and indirectly support cancer survival by interacting with endothelial cells and carcinoma associated fibroblasts. [16] Tregs also have immunosuppressive mechanisms that can make CD8+ cells less effective. [47]

T cells reach tumor sites via the vascular system, where the tumor microenvironment appears to preferentially recruit other immune cells over T cells. One such discriminating mechanism is the release of cell-type specific chemokines. Another is the expression of the apoptosis inducer Fas ligand (FasL) in the vasculature of ovarian, colon, prostate, breast, bladder and renal tumors. Tumors with a high expression of FasL has been shown to contain an abundancy of Tregs, but few CD8+ cells. [3]

T cells must replicate after arriving at the tumor site to effectively kill the cancer cells, survive hostile elements and migrate through the stroma to the cancer cells. This is affected by the tumor microenvironment. The draining lymph nodes are the likely location for cancer specific T cell replication, although this also occurs within the tumor. [3]

Research

Models

Several in vitro and in vivo models have been developed that seek to replicate the TME in a controlled environment. Tumor immortalised cell lines and primary cell cultures have been long used in order to study various tumors. They are quick to set up and inexpensive, but simplistic and prone to genetic drift. [73] 3D tumor models have been developed as a more spatially representative model of the TME. Spheroid cultures, scaffolds and organoids are generally derived from stem cells or ex vivo and are much better at recreating the tumour architecture than 2D cell cultures. [74]

Drug development

Advancements in remodeling nanotherapeutics have led to progress in suppressing cancer metastasis and reducing the likelihood of cancer occurrence. Strategies included regulation of hypoxia, angiogenesis, cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), and tumor-associated macrophages. These approaches aimed to improve anti-tumor effects and sensitize other therapies. [75] Researchers have discovered that the use of ferumoxytol suppress tumor growth by inducing transition of macrophages to proinflammatory types. [76] Nanocarrier vehicles (~20–200 nm in diameter) can transport drugs and other therapeutic molecules. These therapies can be targeted to selectively extravasate through tumor vasculature. [22] [77] These efforts include protein capsids [78] and liposomes. [79] However, as some important, normal tissues, such as the liver and kidneys, also have fenestrated endothelium, the nanocarrier size (10–100 nm, with greater retention in tumors seen in using larger nanocarriers) and charge (anionic or neutral) must be considered. [22] Lymphatic vessels do not usually develop with the tumor, leading to increased interstitial fluid pressure, which may block tumor access. [22] [80]

Therapies

Antibodies

Bevacizumab is clinically approved in the US to treat a variety of cancers by targeting VEGF-A, which is produced by both carcinoma associated fibroblasts and tumor-associated macrophages, thus slowing angiogenesis. It was initially approved for metastatic colorectal cancer, but its uses now span various cancers. [81]

Targeting immunoregulatory membrane receptors succeeded in some patients with melanoma, non-small-cell lung carcinoma, urothelial bladder cancer and renal cell cancer. In mice, anti-CTLA-4 therapy leads to clearance from the tumor of FOXP3 + regulatory T cells (Tregs) whose presence may impair effector T cell function. [82]

Kinase inhibitors

Mutated kinases are common in cancer cells, making them attractive targets for anticancer drugs. Kinase inhibitors are potent, specific and target abnormal kinases while minimizing toxicity. Kinase inhibitors have expanded treatment options for various cancers. [83]

Tyrosine kinase inhibitors (TKIs), such as erlotinib, lapatinib, and gefitinib, target epidermal growth factor receptors (EGFRs) in cancer by blocking the activity of protein tyrosine kinases (PTKs). This show promise in modulating the tumor microenvironment, resulting in cancer regression. Understanding how TKIs modulates the tumor microenvironment may offer another form of cancer treatment. [84] [85]

Chimeric antigen receptor cell therapy

Chimeric antigen receptors (CAR) T cell therapy is an immunotherapy treatment that uses genetically modified T lymphocytes to effectively target tumor cells. CARs are programmed to target tumor-associated antigens as well as replicate rapidly and homogenously, making them potentially very effective as a cancer-therapy. [86] [87] Since the tumor microenvironment has several barriers that limit the ability of CAR T cells to infiltrate the tumor, several strategies have been developed to address this. Localized delivery of CAR T cells in glioblastoma suggested improved anti-tumor activity and engineering these cells to overexpress chemokine receptors suggested improvement of CAR T cell trafficking. [88] As this therapy expands to other diseases, managing its unique toxicity profile, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and cytopenias, becomes increasingly more important. [89]

See also

Related Research Articles

<span class="mw-page-title-main">Metastasis</span> Spread of a disease inside a body

Metastasis is a pathogenic agent's spread from an initial or primary site to a different or secondary site within the host's body; the term is typically used when referring to metastasis by a cancerous tumor. The newly pathological sites, then, are metastases (mets). It is generally distinguished from cancer invasion, which is the direct extension and penetration by cancer cells into neighboring tissues.

<span class="mw-page-title-main">Wound healing</span> Series of events that restore integrity to damaged tissue after an injury

Wound healing refers to a living organism's replacement of destroyed or damaged tissue by newly produced tissue.

Stromal cells, or mesenchymal stromal cells, are differentiating cells found in abundance within bone marrow but can also be seen all around the body. Stromal cells can become connective tissue cells of any organ, for example in the uterine mucosa (endometrium), prostate, bone marrow, lymph node and the ovary. They are cells that support the function of the parenchymal cells of that organ. The most common stromal cells include fibroblasts and pericytes. The term stromal comes from Latin stromat-, "bed covering", and Ancient Greek στρῶμα, strôma, "bed".

Intravasation is the invasion of cancer cells through the basement membrane into a blood or lymphatic vessel. Intravasation is one of several carcinogenic events that initiate the escape of cancerous cells from their primary sites. Other mechanisms include invasion through basement membranes, extravasation, and colonization of distant metastatic sites. Cancer cell chemotaxis also relies on this migratory behavior to arrive at a secondary destination designated for cancer cell colonization.

<span class="mw-page-title-main">Endoglin</span> Protein-coding gene in the species Homo sapiens

Endoglin (ENG) is a type I membrane glycoprotein located on cell surfaces and is part of the TGF beta receptor complex. It is also commonly referred to as CD105, END, FLJ41744, HHT1, ORW and ORW1. It has a crucial role in angiogenesis, therefore, making it an important protein for tumor growth, survival and metastasis of cancer cells to other locations in the body.

<span class="mw-page-title-main">Microvesicle</span> Type of extracellular vesicle

Microvesicles are a type of extracellular vesicle (EV) that are released from the cell membrane. In multicellular organisms, microvesicles and other EVs are found both in tissues and in many types of body fluids. Delimited by a phospholipid bilayer, microvesicles can be as small as the smallest EVs or as large as 1000 nm. They are considered to be larger, on average, than intracellularly-generated EVs known as exosomes. Microvesicles play a role in intercellular communication and can transport molecules such as mRNA, miRNA, and proteins between cells.

<span class="mw-page-title-main">MMP9</span> Protein-coding gene in the species Homo sapiens

Matrix metalloproteinase-9 (MMP-9), also known as 92 kDa type IV collagenase, 92 kDa gelatinase or gelatinase B (GELB), is a matrixin, a class of enzymes that belong to the zinc-metalloproteinases family involved in the degradation of the extracellular matrix. In humans the MMP9 gene encodes for a signal peptide, a propeptide, a catalytic domain with inserted three repeats of fibronectin type II domain followed by a C-terminal hemopexin-like domain.

<span class="mw-page-title-main">MMP2</span> Protein-coding gene in the species Homo sapiens

72 kDa type IV collagenase also known as matrix metalloproteinase-2 (MMP-2) and gelatinase A is an enzyme that in humans is encoded by the MMP2 gene. The MMP2 gene is located on chromosome 16 at position 12.2.

<span class="mw-page-title-main">Periostin</span> Protein-coding gene in the species Homo sapiens

Periostin is a protein that in humans is encoded by the POSTN gene. Periostin functions as a ligand for alpha-V/beta-3 and alpha-V/beta-5 integrins to support adhesion and migration of epithelial cells.

A macrophage-activating factor (MAF) is a lymphokine or other receptor based signal that primes macrophages towards cytotoxicity to tumors, cytokine secretion, or clearance of pathogens. Similar molecules may cause development of an inhibitory, regulatory phenotype. A MAF can also alter the ability of macrophages to present MHC I antigen, participate in Th responses, and/or affect other immune responses.

<span class="mw-page-title-main">Desmoplasia</span> Growth of fibrous or connective tissue

In medicine, desmoplasia is the growth of fibrous connective tissue. It is also called a desmoplastic reaction to emphasize that it is secondary to an insult. Desmoplasia may occur around a neoplasm, causing dense fibrosis around the tumor, or scar tissue (adhesions) within the abdomen after abdominal surgery.

Angiogenesis is the process of forming new blood vessels from existing blood vessels, formed in vasculogenesis. It is a highly complex process involving extensive interplay between cells, soluble factors, and the extracellular matrix (ECM). Angiogenesis is critical during normal physiological development, but it also occurs in adults during inflammation, wound healing, ischemia, and in pathological conditions such as rheumatoid arthritis, hemangioma, and tumor growth. Proteolysis has been indicated as one of the first and most sustained activities involved in the formation of new blood vessels. Numerous proteases including matrix metalloproteinases (MMPs), a disintegrin and metalloproteinase domain (ADAM), a disintegrin and metalloproteinase domain with throbospondin motifs (ADAMTS), and cysteine and serine proteases are involved in angiogenesis. This article focuses on the important and diverse roles that these proteases play in the regulation of angiogenesis.

<span class="mw-page-title-main">Metastatic breast cancer</span> Type of cancer

Metastatic breast cancer, also referred to as metastases, advanced breast cancer, secondary tumors, secondaries or stage IV breast cancer, is a stage of breast cancer where the breast cancer cells have spread to distant sites beyond the axillary lymph nodes. There is no cure for metastatic breast cancer; there is no stage after IV.

Tumor-associated macrophages (TAMs) are a class of immune cells present in high numbers in the microenvironment of solid tumors. They are heavily involved in cancer-related inflammation. Macrophages are known to originate from bone marrow-derived blood monocytes or yolk sac progenitors, but the exact origin of TAMs in human tumors remains to be elucidated. The composition of monocyte-derived macrophages and tissue-resident macrophages in the tumor microenvironment depends on the tumor type, stage, size, and location, thus it has been proposed that TAM identity and heterogeneity is the outcome of interactions between tumor-derived, tissue-specific, and developmental signals.

<span class="mw-page-title-main">Vasculogenic mimicry</span>

Vasculogenic mimicry (VM) is a strategy used by tumors to ensure sufficient blood supply is brought to its cells through establishing new tumor vascularization. This process is similar to tumor angiogenesis; on the other hand vascular mimicry is unique in that this process occurs independent of endothelial cells. Vasculature is instead developed de novo by cancer cells, which under stress conditions such as hypoxia, express similar properties to stem cells, capable of differentiating to mimic the function of endothelial cells and form vasculature-like structures. The ability of tumors to develop and harness nearby vasculature is considered one of the hallmarks of cancer disease development and is thought to be closely linked to tumor invasion and metastasis. Vascular mimicry has been observed predominantly in aggressive and metastatic cancers and has been associated with negative tumor characteristics such as increased metastasis, increased tissue invasion, and overall poor outcomes for patient survival. Vascular mimicry poses a serious problem for current therapeutic strategies due to its ability to function in the presence of Anti-angiogenic therapeutic agents. In fact, such therapeutics have been found to actually drive VM formation in tumors, causing more aggressive and difficult to treat tumors to develop.

<span class="mw-page-title-main">Tumor-associated endothelial cell</span>

Tumor-associated endothelial cells or tumor endothelial cells (TECs) refers to cells lining the tumor-associated blood vessels that control the passage of nutrients into surrounding tumor tissue. Across different cancer types, tumor-associated blood vessels have been discovered to differ significantly from normal blood vessels in morphology, gene expression, and functionality in ways that promote cancer progression. There has been notable interest in developing cancer therapeutics that capitalize on these abnormalities of the tumor-associated endothelium to destroy tumors.

A pre-metastatic niche is an environment in a secondary organ that can be conducive to the metastasis of a primary tumor. Such a niche provides favorable conditions for growth, and eventual metastasis, in an otherwise foreign and hostile environment for the primary tumor cells. This concept demonstrated the fundamental role of the microenvironment in regulating tumor growth and metastasis. The discovery of the pre-metastatic niche has fostered new research regarding the potential treatment of metastases, including targeting myeloid derived suppressor cells, and stromal cell plasticity including fibroblasts and pericytes and perivascular smooth muscle cells and (attempts to stop the flow of vesicles from primary tumors to pre-metastatic niches in secondary organs and different combinations of microenvironment targeted therapies.

A cancer-associated fibroblast (CAF) is a cell type within the tumor microenvironment that promotes tumorigenic features by initiating the remodelling of the extracellular matrix or by secreting cytokines. CAFs are a complex and abundant cell type within the tumour microenvironment; the number cannot decrease, as they are unable to undergo apoptosis.

The host response to cancer therapy is defined as a physiological response of the non-malignant cells of the body to a specific cancer therapy. The response is therapy-specific, occurring independently of cancer type or stage.

<span class="mw-page-title-main">Michele De Palma</span> Italian biologist

Michele 'Miki' De Palma is an Italian biologist and a professor at EPFL. He is known for his work on the role of macrophages in cancer progression and the discovery of Tie2-expressing angiogenic monocytes.

References

  1. Piñeiro Fernández J, Luddy KA, Harmon C, O'Farrelly C (2019). "Hepatic Tumor Microenvironments and Effects on NK Cell Phenotype and Function". International Journal of Molecular Sciences. 20 (17): 4131. doi: 10.3390/ijms20174131 . ISSN   1422-0067. PMC   6747260 . PMID   31450598.
  2. 1 2 Alfarouk KO, Muddathir AK, Shayoub ME (January 2011). "Tumor acidity as evolutionary spite". Cancers. 3 (1): 408–414. doi: 10.3390/cancers3010408 . PMC   3756368 . PMID   24310355.
  3. 1 2 3 Joyce JA, Fearon DT (April 2015). "T cell exclusion, immune privilege, and the tumor microenvironment". Science. 348 (6230): 74–80. Bibcode:2015Sci...348...74J. doi: 10.1126/science.aaa6204 . PMID   25838376.
  4. 1 2 Spill F, Reynolds DS, Kamm RD, Zaman MH (August 2016). "Impact of the physical microenvironment on tumor progression and metastasis". Current Opinion in Biotechnology. 40: 41–48. doi:10.1016/j.copbio.2016.02.007. PMC   4975620 . PMID   26938687.
  5. 1 2 3 4 Weber CE, Kuo PC (September 2012). "The tumor microenvironment". Surgical Oncology. 21 (3): 172–177. doi:10.1016/j.suronc.2011.09.001. PMID   21963199.
  6. Korneev KV, Atretkhany KN, Drutskaya MS, Grivennikov SI, Kuprash DV, Nedospasov SA (January 2017). "TLR-signaling and proinflammatory cytokines as drivers of tumorigenesis". Cytokine. 89: 127–135. doi:10.1016/j.cyto.2016.01.021. PMID   26854213.
  7. Ghoshdastider U, Rohatgi N, Mojtabavi Naeini M, Baruah P, Revkov E, Guo YA, et al. (April 2021). "Pan-Cancer Analysis of Ligand-Receptor Cross-talk in the Tumor Microenvironment". Cancer Research. 81 (7): 1802–1812. doi: 10.1158/0008-5472.CAN-20-2352 . PMID   33547160. S2CID   232432582.
  8. Žavbi J, Breznik B (2021). "Modelling the microenvironment of the most aggressive brain tumours for preclinical studies". Advances in Cancer Biology - Metastasis. 3: 100017. doi: 10.1016/j.adcanc.2021.100017 . ISSN   2667-3940. S2CID   244452599.
  9. Xinyuan T, Lei Y, Jianping S, Rongwei Z, Ruiwen S, Ye Z, et al. (2023-10-01). "Advances in the role of gut microbiota in the regulation of the tumor microenvironment (Review)". Oncology Reports. 50 (4): 1–15. doi:10.3892/or.2023.8618. ISSN   1021-335X. PMC   10485805 . PMID   37615187.
  10. 1 2 3 Akhtar M, Haider A, Rashid S, Al-Nabet AD (January 2019). "Paget's "Seed and Soil" Theory of Cancer Metastasis: An Idea Whose Time has Come". Advances in Anatomic Pathology. 26 (1): 69–74. doi:10.1097/PAP.0000000000000219. ISSN   1072-4109. PMID   30339548. S2CID   53010974.
  11. 1 2 3 4 5 Mao Y, Keller ET, Garfield DH, Shen K, Wang J (2013-06-01). "Stromal cells in tumor microenvironment and breast cancer". Cancer and Metastasis Reviews. 32 (1): 303–315. doi:10.1007/s10555-012-9415-3. ISSN   1573-7233. PMC   4432936 . PMID   23114846.
  12. Liu Q, Zhang H, Jiang X, Qian C, Liu Z, Luo D (2017-12-02). "Factors involved in cancer metastasis: a better understanding to "seed and soil" hypothesis". Molecular Cancer. 16 (1): 176. doi: 10.1186/s12943-017-0742-4 . ISSN   1476-4598. PMC   5712107 . PMID   29197379.
  13. da Costa VR, Araldi RP, Vigerelli H, D'Ámelio F, Mendes TB, Gonzaga V, et al. (October 2021). "Exosomes in the Tumor Microenvironment: From Biology to Clinical Applications". Cells. 10 (10): 2617. doi: 10.3390/cells10102617 . ISSN   2073-4409. PMC   8533895 . PMID   34685596.
  14. Maman S, Witz IP (June 2018). "A history of exploring cancer in context". Nature Reviews Cancer. 18 (6): 359–376. doi:10.1038/s41568-018-0006-7. ISSN   1474-1768. PMID   29700396. S2CID   13777357.
  15. Al-Ostoot FH, Salah S, Khamees HA, Khanum SA (2021-01-01). "Tumor angiogenesis: Current challenges and therapeutic opportunities". Cancer Treatment and Research Communications. 28: 100422. doi: 10.1016/j.ctarc.2021.100422 . ISSN   2468-2942. PMID   34147821.
  16. 1 2 3 4 5 6 7 8 9 10 11 Anderson NM, Simon MC (August 2020). "The tumor microenvironment". Current Biology. 30 (16): R921–R925. Bibcode:2020CBio...30.R921A. doi:10.1016/j.cub.2020.06.081. ISSN   0960-9822. PMC   8194051 . PMID   32810447.
  17. Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, et al. (2020-09-30). "The role of microenvironment in tumor angiogenesis". Journal of Experimental & Clinical Cancer Research. 39 (1): 204. doi: 10.1186/s13046-020-01709-5 . ISSN   1756-9966. PMC   7526376 . PMID   32993787.
  18. 1 2 3 Li Y, Zhao L, Li XF (2021-08-05). "Hypoxia and the Tumor Microenvironment". Technology in Cancer Research & Treatment. 20: 15330338211036304. doi:10.1177/15330338211036304. ISSN   1533-0346. PMC   8358492 . PMID   34350796.
  19. Bhat SM, Badiger VA, Vasishta S, Chakraborty J, Prasad S, Ghosh S, et al. (2021-12-01). "3D tumor angiogenesis models: recent advances and challenges". Journal of Cancer Research and Clinical Oncology. 147 (12): 3477–3494. doi:10.1007/s00432-021-03814-0. ISSN   1432-1335. PMC   8557138 . PMID   34613483.
  20. Hanahan D, Monje M (2023-03-13). "Cancer hallmarks intersect with neuroscience in the tumor microenvironment". Cancer Cell. 41 (3): 573–580. doi:10.1016/j.ccell.2023.02.012. ISSN   1535-6108. PMC   10202656 . PMID   36917953.
  21. Alasvand N, Urbanska AM, Rahmati M, Saeidifar M, Gungor-Ozkerim PS, Sefat F, et al. (2017-01-01), Grumezescu AM (ed.), "Chapter 13 - Therapeutic Nanoparticles for Targeted Delivery of Anticancer Drugs", Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics, Elsevier, pp. 245–259, doi:10.1016/b978-0-323-52725-5.00013-7, ISBN   978-0-323-52725-5 , retrieved 2024-02-19
  22. 1 2 3 4 Danhier F, Feron O, Préat V (December 2010). "To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery". Journal of Controlled Release. 148 (2): 135–146. doi:10.1016/j.jconrel.2010.08.027. PMID   20797419.
  23. Tang M, Bolderson E, O'Byrne KJ, Richard DJ (2021). "Tumor Hypoxia Drives Genomic Instability". Frontiers in Cell and Developmental Biology. 9. doi: 10.3389/fcell.2021.626229 . ISSN   2296-634X. PMC   8007910 . PMID   33796526.
  24. Wicks EE, Semenza GL (2022-06-01). "Hypoxia-inducible factors: cancer progression and clinical translation". The Journal of Clinical Investigation. 132 (11). doi:10.1172/JCI159839. ISSN   0021-9738. PMC   9151701 . PMID   35642641.
  25. 1 2 3 4 Chen G, Wu K, Li H, Xia D, He T (2022). "Role of hypoxia in the tumor microenvironment and targeted therapy". Frontiers in Oncology. 12. doi: 10.3389/fonc.2022.961637 . ISSN   2234-943X. PMC   9545774 . PMID   36212414.
  26. Lee SH, Griffiths JR (June 2020). "How and Why Are Cancers Acidic? Carbonic Anhydrase IX and the Homeostatic Control of Tumour Extracellular pH". Cancers. 12 (6): 1616. doi: 10.3390/cancers12061616 . PMC   7352839 . PMID   32570870.
  27. 1 2 Werb Z, Lu P (July–August 2015). "The Role of Stroma in Tumor Development". The Cancer Journal. 21 (4): 250–253. doi:10.1097/PPO.0000000000000127. ISSN   1540-336X. PMC   4963227 . PMID   26222075.
  28. 1 2 3 4 Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, et al. (2023-09-04). "Stromal cells in the tumor microenvironment: accomplices of tumor progression?". Cell Death & Disease. 14 (9): 587. doi:10.1038/s41419-023-06110-6. ISSN   2041-4889. PMC   10477351 . PMID   37666813.
  29. Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C (2021-07-11). "Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy?". Cancers. 13 (14): 3466. doi: 10.3390/cancers13143466 . ISSN   2072-6694. PMC   8303391 . PMID   34298680.
  30. Arora L, Pal D (2021). "Remodeling of Stromal Cells and Immune Landscape in Microenvironment During Tumor Progression". Frontiers in Oncology. 11. doi: 10.3389/fonc.2021.596798 . ISSN   2234-943X. PMC   7982455 . PMID   33763348.
  31. 1 2 Glabman RA, Choyke PL, Sato N (January 2022). "Cancer-Associated Fibroblasts: Tumorigenicity and Targeting for Cancer Therapy". Cancers. 14 (16): 3906. doi: 10.3390/cancers14163906 . ISSN   2072-6694. PMC   9405783 . PMID   36010899.
  32. Yang D, Liu J, Qian H, Zhuang Q (July 2023). "Cancer-associated fibroblasts: from basic science to anticancer therapy". Experimental & Molecular Medicine. 55 (7): 1322–1332. doi:10.1038/s12276-023-01013-0. ISSN   2092-6413. PMC   10394065 . PMID   37394578.
  33. 1 2 3 Ping Q, Yan R, Cheng X, Wang W, Zhong Y, Hou Z, et al. (September 2021). "Cancer-associated fibroblasts: overview, progress, challenges, and directions". Cancer Gene Therapy. 28 (9): 984–999. doi:10.1038/s41417-021-00318-4. ISSN   1476-5500. PMID   33712707.
  34. 1 2 Cords L, Tietscher S, Anzeneder T, Langwieder C, Rees M, de Souza N, et al. (2023-07-18). "Cancer-associated fibroblast classification in single-cell and spatial proteomics data". Nature Communications. 14 (1): 4294. Bibcode:2023NatCo..14.4294C. doi:10.1038/s41467-023-39762-1. ISSN   2041-1723. PMC   10354071 . PMID   37463917.
  35. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. (March 2020). "A framework for advancing our understanding of cancer-associated fibroblasts". Nature Reviews Cancer. 20 (3): 174–186. doi:10.1038/s41568-019-0238-1. ISSN   1474-1768. PMC   7046529 . PMID   31980749.
  36. Bonnans C, Chou J, Werb Z (2015-02-04). "Remodelling the extracellular matrix in development and disease". Nature Reviews Molecular Cell Biology. 15 (12): 786–801. doi:10.1038/nrm3904. ISSN   1471-0080. PMC   4316204 . PMID   25415508.
  37. Fromme JE, Zigrino P (2022). "The Role of Extracellular Matrix Remodeling in Skin Tumor Progression and Therapeutic Resistance". Frontiers in Molecular Biosciences. 9. doi: 10.3389/fmolb.2022.864302 . ISSN   2296-889X. PMC   9086898 . PMID   35558554.
  38. Diller RB, Tabor AJ (2022-07-01). "The Role of the Extracellular Matrix (ECM) in Wound Healing: A Review". Biomimetics. 7 (3): 87. doi: 10.3390/biomimetics7030087 . ISSN   2313-7673. PMC   9326521 . PMID   35892357.
  39. Brassart-Pasco S, Brézillon S, Brassart B, Ramont L, Oudart JB, Monboisse JC (2020). "Tumor Microenvironment: Extracellular Matrix Alterations Influence Tumor Progression". Frontiers in Oncology. 10: 397. doi: 10.3389/fonc.2020.00397 . ISSN   2234-943X. PMC   7174611 . PMID   32351878.
  40. 1 2 Mohan V, Das A, Sagi I (2020-05-01). "Emerging roles of ECM remodeling processes in cancer". Seminars in Cancer Biology. Translating Extracellular Matrix. 62: 192–200. doi:10.1016/j.semcancer.2019.09.004. ISSN   1044-579X. PMID   31518697. S2CID   202571768.
  41. Larsen M, Artym VV, Green JA, Yamada KM (2006-10-01). "The matrix reorganized: extracellular matrix remodeling and integrin signaling". Current Opinion in Cell Biology. Cell-to-cell contact and extracellular matrix. 18 (5): 463–471. doi:10.1016/j.ceb.2006.08.009. ISSN   0955-0674. PMID   16919434.
  42. Kai F, Drain AP, Weaver VM (2019-05-06). "The Extracellular Matrix Modulates the Metastatic Journey". Developmental Cell. 49 (3): 332–346. doi:10.1016/j.devcel.2019.03.026. ISSN   1878-1551. PMC   6527347 . PMID   31063753.
  43. 1 2 Dzobo K, Dandara C (2023-04-05). "The Extracellular Matrix: Its Composition, Function, Remodeling, and Role in Tumorigenesis". Biomimetics. 8 (2): 146. doi: 10.3390/biomimetics8020146 . ISSN   2313-7673. PMC   10123695 . PMID   37092398.
  44. Giussani M, Triulzi T, Sozzi G, Tagliabue E (2019-01-23). "Tumor Extracellular Matrix Remodeling: New Perspectives as a Circulating Tool in the Diagnosis and Prognosis of Solid Tumors". Cells. 8 (2): 81. doi: 10.3390/cells8020081 . ISSN   2073-4409. PMC   6406979 . PMID   30678058.
  45. Wullkopf L, West AK, Leijnse N, Cox TR, Madsen CD, Oddershede LB, et al. (October 2018). Discher D (ed.). "Cancer cells' ability to mechanically adjust to extracellular matrix stiffness correlates with their invasive potential". Molecular Biology of the Cell. 29 (20): 2378–2385. doi:10.1091/mbc.E18-05-0319. ISSN   1059-1524. PMC   6233061 . PMID   30091653.
  46. Popova NV, Jücker M (January 2022). "The Functional Role of Extracellular Matrix Proteins in Cancer". Cancers. 14 (1): 238. doi: 10.3390/cancers14010238 . ISSN   2072-6694. PMC   8750014 . PMID   35008401.
  47. 1 2 3 4 5 6 7 8 Lei X, Lei Y, Li JK, Du WX, Li RG, Yang J, et al. (2020-02-01). "Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy". Cancer Letters. 470: 126–133. doi:10.1016/j.canlet.2019.11.009. ISSN   0304-3835. PMID   31730903. S2CID   208063582.
  48. Mantovani A, Allavena P, Sica A, Balkwill F (July 2008). "Cancer-related inflammation". Nature. 454 (7203): 436–444. Bibcode:2008Natur.454..436M. doi:10.1038/nature07205. hdl: 2434/145688 . PMID   18650914. S2CID   4429118.
  49. 1 2 Mathias RA, Gopal SK, Simpson RJ (January 2013). "Contribution of cells undergoing epithelial-mesenchymal transition to the tumour microenvironment". Journal of Proteomics. 78: 545–557. doi:10.1016/j.jprot.2012.10.016. PMID   23099347.
  50. Balkwill F, Charles KA, Mantovani A (March 2005). "Smoldering and polarized inflammation in the initiation and promotion of malignant disease". Cancer Cell. 7 (3): 211–217. doi: 10.1016/j.ccr.2005.02.013 . PMID   15766659.
  51. Biswas SK, Gangi L, Paul S, Schioppa T, Saccani A, Sironi M, et al. (March 2006). "A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation)". Blood. 107 (5): 2112–2122. doi: 10.1182/blood-2005-01-0428 . PMID   16269622. S2CID   5884781.
  52. Qian BZ, Pollard JW (April 2010). "Macrophage diversity enhances tumor progression and metastasis". Cell. 141 (1): 39–51. doi:10.1016/j.cell.2010.03.014. PMC   4994190 . PMID   20371344.
  53. Zhang W, Wang L, Zhou D, Cui Q, Zhao D, Wu Y (January 2011). "Expression of tumor-associated macrophages and vascular endothelial growth factor correlates with poor prognosis of peripheral T-cell lymphoma, not otherwise specified". Leukemia & Lymphoma. 52 (1): 46–52. doi:10.3109/10428194.2010.529204. PMID   21077742. S2CID   26116121.
  54. Zhang BC, Gao J, Wang J, Rao ZG, Wang BC, Gao JF (December 2011). "Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma". Medical Oncology. 28 (4): 1447–1452. doi:10.1007/s12032-010-9638-5. PMID   20676804. S2CID   24840259.
  55. Yang M, Chen J, Su F, Yu B, Su F, Lin L, et al. (September 2011). "Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells". Molecular Cancer. 10 (117): 117. doi: 10.1186/1476-4598-10-117 . PMC   3190352 . PMID   21939504.
  56. Coffelt SB, Wellenstein MD, de Visser KE (July 2016). "Neutrophils in cancer: neutral no more" (PDF). Nature Reviews. Cancer. 16 (7): 431–446. doi:10.1038/nrc.2016.52. PMID   27282249. S2CID   4393159.
  57. Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, et al. (August 2015). "The prognostic landscape of genes and infiltrating immune cells across human cancers". Nature Medicine. 21 (8): 938–945. doi:10.1038/nm.3909. PMC   4852857 . PMID   26193342.
  58. Engblom C, Pfirschke C, Pittet MJ (July 2016). "The role of myeloid cells in cancer therapies". Nature Reviews. Cancer. 16 (7): 447–462. doi:10.1038/nrc.2016.54. PMID   27339708. S2CID   21924175.
  59. Huang SH, Waldron JN, Milosevic M, Shen X, Ringash J, Su J, et al. (February 2015). "Prognostic value of pretreatment circulating neutrophils, monocytes, and lymphocytes in oropharyngeal cancer stratified by human papillomavirus status". Cancer. 121 (4): 545–555. doi: 10.1002/cncr.29100 . PMID   25336438. S2CID   926930.
  60. Jiang L, Jiang S, Situ D, Lin Y, Yang H, Li Y, et al. (April 2015). "Prognostic value of monocyte and neutrophils to lymphocytes ratio in patients with metastatic soft tissue sarcoma". Oncotarget. 6 (11): 9542–9550. doi:10.18632/oncotarget.3283. PMC   4496237 . PMID   25865224.
  61. Wu WC, Sun HW, Chen HT, Liang J, Yu XJ, Wu C, et al. (March 2014). "Circulating hematopoietic stem and progenitor cells are myeloid-biased in cancer patients". Proceedings of the National Academy of Sciences of the United States of America. 111 (11): 4221–4226. Bibcode:2014PNAS..111.4221W. doi: 10.1073/pnas.1320753111 . PMC   3964061 . PMID   24591638.
  62. Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, et al. (June 2015). "IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis". Nature. 522 (7556): 345–348. Bibcode:2015Natur.522..345C. doi:10.1038/nature14282. PMC   4475637 . PMID   25822788.
  63. Engblom C, Pfirschke C, Zilionis R, Da Silva Martins J, Bos SA, Courties G, et al. (December 2017). "Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neutrophils". Science. 358 (6367): eaal5081. doi:10.1126/science.aal5081. PMC   6343476 . PMID   29191879.
  64. Casbon AJ, Reynaud D, Park C, Khuc E, Gan DD, Schepers K, et al. (February 2015). "Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils". Proceedings of the National Academy of Sciences of the United States of America. 112 (6): E566–E575. Bibcode:2015PNAS..112E.566C. doi: 10.1073/pnas.1424927112 . PMC   4330753 . PMID   25624500.
  65. Wculek SK, Malanchi I (December 2015). "Neutrophils support lung colonization of metastasis-initiating breast cancer cells". Nature. 528 (7582): 413–417. Bibcode:2015Natur.528..413W. doi:10.1038/nature16140. PMC   4700594 . PMID   26649828.
  66. Finisguerra V, Di Conza G, Di Matteo M, Serneels J, Costa S, Thompson AA, et al. (June 2015). "MET is required for the recruitment of anti-tumoural neutrophils". Nature. 522 (7556): 349–353. Bibcode:2015Natur.522..349F. doi:10.1038/nature14407. PMC   4594765 . PMID   25985180.
  67. Granot Z, Henke E, Comen EA, King TA, Norton L, Benezra R (September 2011). "Tumor entrained neutrophils inhibit seeding in the premetastatic lung". Cancer Cell. 20 (3): 300–314. doi:10.1016/j.ccr.2011.08.012. PMC   3172582 . PMID   21907922.
  68. "tumor-infiltrating lymphocyte". www.cancer.gov. 2011-02-02. Retrieved 2024-02-19.
  69. 1 2 3 Whiteside TL (2022). "Tumor-Infiltrating Lymphocytes and Their Role in Solid Tumor Progression". Interaction of Immune and Cancer Cells. Experientia Supplementum. Vol. 113. pp. 89–106. doi:10.1007/978-3-030-91311-3_3. ISBN   978-3-030-91310-6. ISSN   1664-431X. PMC   9113058 . PMID   35165861.
  70. Turcotte S, Rosenberg SA (2011). "Immunotherapy for metastatic solid cancers". Advances in Surgery. 45: 341–360. doi:10.1016/j.yasu.2011.04.003. PMC   3578602 . PMID   21954698.
  71. Valenti R, Huber V, Iero M, Filipazzi P, Parmiani G, Rivoltini L (April 2007). "Tumor-released microvesicles as vehicles of immunosuppression". Cancer Research. 67 (7): 2912–2915. doi: 10.1158/0008-5472.CAN-07-0520 . PMID   17409393.
  72. Clayton A, Tabi Z (May–June 2005). "Exosomes and the MICA-NKG2D system in cancer". Blood Cells, Molecules & Diseases. 34 (3): 206–213. doi:10.1016/j.bcmd.2005.03.003. PMID   15885603.
  73. Freedman LP, Gibson MC, Ethier SP, Soule HR, Neve RM, Reid YA (June 2015). "Reproducibility: changing the policies and culture of cell line authentication". Nature Methods. 12 (6): 493–497. doi:10.1038/nmeth.3403. PMID   26020501. S2CID   20557369.
  74. Franchi-Mendes T, Eduardo R, Domenici G, Brito C (September 2021). "3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment". Cancers. 13 (18): 4610. doi: 10.3390/cancers13184610 . PMC   8468887 . PMID   34572836.
  75. Feng Y, Liao Z, Zhang H, Xie X, You F, Liao X, et al. (January 2023). "Emerging nanomedicines strategies focused on tumor microenvironment against cancer recurrence and metastasis". Chemical Engineering Journal. 452: 139506. Bibcode:2023ChEnJ.45239506F. doi:10.1016/j.cej.2022.139506. S2CID   252676223.
  76. Raju GS, Pavitra E, Varaprasad GL, Bandaru SS, Nagaraju GP, Farran B, et al. (June 2022). "Nanoparticles mediated tumor microenvironment modulation: current advances and applications". Journal of Nanobiotechnology. 20 (1): 274. doi: 10.1186/s12951-022-01476-9 . PMC   9195263 . PMID   35701781.
  77. Unezaki S, Maruyama K, Hosoda JI, Nagae I, Koyanagi Y, Nakata M, et al. (22 November 1996). "Direct measurement of the extravasation of polyethyleneglycol-coated liposomes into solid tumor tissue by in vivo fluorescence microscopy". International Journal of Pharmaceutics. 144 (1): 11–17. doi:10.1016/S0378-5173(96)04674-1.
  78. Lilavivat S, Sardar D, Jana S, Thomas GC, Woycechowsky KJ (August 2012). "In vivo encapsulation of nucleic acids using an engineered nonviral protein capsid". Journal of the American Chemical Society. 134 (32): 13152–13155. doi:10.1021/ja302743g. PMID   22827162.
  79. Ramishetti S, Huang L (December 2012). "Intelligent design of multifunctional lipid-coated nanoparticle platforms for cancer therapy". Therapeutic Delivery. 3 (12): 1429–1445. doi:10.4155/tde.12.127. PMC   3584330 . PMID   23323560.
  80. Jain RK (June 1987). "Transport of molecules in the tumor interstitium: a review". Cancer Research. 47 (12): 3039–3051. PMID   3555767.
  81. Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, et al. (June 2020). "Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook". Cancer Treatment Reviews. 86: 102017. doi: 10.1016/j.ctrv.2020.102017 . ISSN   0305-7372. PMID   32335505.
  82. Li C, Jiang P, Wei S, Xu X, Wang J (July 2020). "Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects". Molecular Cancer. 19 (1): 116. doi: 10.1186/s12943-020-01234-1 . PMC   7367382 . PMID   32680511.
  83. "Protein Kinase Inhibitors", LiverTox: Clinical and Research Information on Drug-Induced Liver Injury, Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases, 2012, PMID   31643906 , retrieved 2024-02-20
  84. Tan HY, Wang N, Lam W, Guo W, Feng Y, Cheng YC (2018-02-19). "Targeting tumour microenvironment by tyrosine kinase inhibitor". Molecular Cancer. 17 (1): 43. doi: 10.1186/s12943-018-0800-6 . ISSN   1476-4598. PMC   5817793 . PMID   29455663.
  85. Mansour MA, Caputo VS, Aleem E (2021-11-01). "Highlights on selected growth factors and their receptors as promising anticancer drug targets". The International Journal of Biochemistry & Cell Biology. 140: 106087. doi:10.1016/j.biocel.2021.106087. ISSN   1357-2725. PMID   34563698. S2CID   237943611.
  86. Kankeu Fonkoua LA, Sirpilla O, Sakemura R, Siegler EL, Kenderian SS (2022-06-16). "CAR T cell therapy and the tumor microenvironment: Current challenges and opportunities". Molecular Therapy - Oncolytics. 25: 69–77. doi:10.1016/j.omto.2022.03.009. ISSN   2372-7705. PMC   8980704 . PMID   35434273.
  87. Zhang C, Durer S, Thandra KC, Kasi A (2024), "Chimeric Antigen Receptor T-Cell Therapy", StatPearls, Treasure Island (FL): StatPearls Publishing, PMID   30725979 , retrieved 2024-02-20
  88. Brown CE, Aguilar B, Starr R, Yang X, Chang WC, Weng L, et al. (January 2018). "Optimization of IL13Rα2-Targeted Chimeric Antigen Receptor T Cells for Improved Anti-tumor Efficacy against Glioblastoma". Molecular Therapy. 26 (1): 31–44. doi: 10.1016/j.ymthe.2017.10.002 . PMC   5763077 . PMID   29103912.
  89. Schubert ML, Schmitt M, Wang L, Ramos CA, Jordan K, Müller-Tidow C, et al. (2021-01-01). "Side-effect management of chimeric antigen receptor (CAR) T-cell therapy". Annals of Oncology. 32 (1): 34–48. doi: 10.1016/j.annonc.2020.10.478 . ISSN   0923-7534. PMID   33098993.