Abstract
Chronic inflammation caused by persistent infections and metabolic disorders is thought to contribute to the increased cancer risk and the accelerated cancer progression. Oppositely, acute inflammation induced by bacteria-based vaccines or that is occurring after cancer selectively inhibits cancer progression and metastasis. However, the interaction between inflammation and cancer may be more complex than the current explanations for the relationship between chronic and acute inflammation and cancer. In this review, we described the impact of inflammation on cancer on the basis of three perspectives, including inflammation with different durations (chronic and acute inflammation), different scopes (systemic and local inflammation) and different occurrence sequences (inflammation occurring after and before cancer). In addition, we also introduced bacteria/virus-based cancer immunotherapies. We perceive that inflammation may be a double-edged sword with cancer-promoting and cancer-suppressing functions in certain cases. We expect to further improve the understanding of the relationship between inflammation and cancer and provide a theoretical basis for further research on their complex interaction.
Keywords: Cancer, Duration, Inflammation, Scope, Sequence
Introduction
In 1828, Jean Nicolas Marjolin observed that squamous cell carcinoma developed around a post-traumatic scar tissue, suggesting that chronic inflammation was involved in cancer progression.1 In 1863, Rudolf Virchow observed leucocytes in neoplastic tissues, which impelled researchers to associate inflammation with cancer.2 Subsequently, Harold F Dvorak reported that cancer and inflammation shared similar signaling pathways, such as cell proliferation, survival, migration and altered angiogenesis, which were regulated by growth factors, pro-inflammatory cytokines and proangiogenic factors.3 In recent years, epidemiological studies have shown that chronic inflammation increases the incidence of various cancers, such as liver, gastric, bladder, cervical and thyroid cancers. The use of non-steroidal anti-inflammatory drugs can reduce the risk and mortality of patients with certain cancers (such as colon and breast cancers).4,5
Although most evidence supports the promoting effect of inflammation in cancer, inflammation exerts anti-cancer effects in some cases. Acute inflammation is usually called ‘therapeutic inflammation’, wherein bacteria- and vaccines-induced immune response exerts an anti-cancer function.6, 7, 8, 9 Furthermore, local inflammation in patients with cancer leads to a better prognosis.10, 11, 12 The above-mentioned studies indicate that inflammation exerts positive or negative effects on cancer based on the duration, scope and sequence of inflammation. Therefore, in this study, we described the impact of inflammation on cancer on the basis of its durations, scopes and occurrence sequences. Meanwhile, we also introduced bacteria/virus-based cancer immunotherapies.
Impact of inflammation with different durations on cancer
Inflammation is a normal pathological process that may occur in various parts of body. In the presence of a pathogen or physical or chemical stimulation, acute inflammation occurs locally to reduce damage at the site, re-establish homeostasis and support wound healing.13 In the acute phase, blood flow and vascular permeability increase with the accumulation of inflammatory mediators and white blood cells.14 Correspondingly, chronic inflammation often occurs without definite signs and symptoms and is often neglected until the disease becomes clinically apparent.15 In both acute and chronic inflammation, the resident cells (such as tissue-resident macrophages; fibroblasts and stromal, endothelial, dendritic and mass cells) are activated first by various soluble factors to initiate an immune response.16 Inflammatory agents such as cytokines, chemokines, histamines and lipid mediators trigger the migration and infiltration of inflammatory cells (e.g., natural killer [NK] cells, lymphocytes, monocytes and neutrophils), which may cause a systemic response (such as leucocytosis, fever, hypotension and cachexia).17 Acute and chronic inflammation frequently share a similar response that involves various immune cells and soluble mediators, but their effect on cancer is distinctly different.
Chronic inflammation in cancer: always the evil
Chronic unresolved inflammation is reported to mediate the initiation and development of cancer.18, 19, 20 Recently, approximately 25% of cancer cases have been estimated to be related to chronic inflammation.21 Although cancer is multi-factorial in origin, various epidemiological and experimental studies suggest that some factor-induced chronic inflammatory conditions may increase the risk of cancer (Table 1):
-
1)
Chronic inflammation caused by persistent infections from viruses, bacteria, nematodes and other micro-organisms contributes to the initiation and development of cancers.22, 23, 24 In 2018, infections constituted 13% of new cancer cases worldwide, and more than one-third of infection-related cancer cases were reported in China.25 Infections with hepatitis B virus (HBV), hepatitis C virus (HCV), human papilloma virus (HPV) and Epstein–Barr virus (EBV) may lead to cancers such as liver and cervical cancers and nasopharyngeal carcinoma.26, 27, 28 Approximately 80% of cases of hepatocellular carcinoma (HCC) are attributed to HBV and HCV infections.29 More than 50% of HCC cases are reported in China, and more than 80% can be attributed to HBV infection.30 In addition, HCC infection predominates in Africa and Asia, where HBV and HCV infections constitute approximately 60% and 20% of etiological factors of HCC, respectively.31 Low incidence rates are reported in the United States, Europe, Egypt and Japan, where HBV and HCV infections are responsible for approximately 20% and more than 60% of HCC cases, respectively.31 Although the mechanism remains uncertain, the HBV gene product HBV X protein (HBx) is confirmed to contribute to the viral pathogenesis and carcinogenesis of HCC by modulating the activities of several signaling pathways, such as interleukin (IL)-6/signal transducer and activator of transcription (STAT) 3, Wnt/-catenin, mitogen-activated protein kinases (MAPK) and nuclear factor kappa-B (NF-B) pathways.32,33 Cervical cancer ranks fourth in terms of global female cancer incidence and mortality; it is mostly caused by HPV infection.34 Approximately 90% of HPV infections regress spontaneously within 1–2 years, and HPV vaccination and screening can reduce HPV-related cervical cancer by more than 90%.35 However, persistent HPV infections (mainly HPV 16 and 18) may lead to cancer.36 Pathways such as Wnt/-catenin, phosphatidylinositol-3-kinase (PI3K)/serine–threonine kinase (Akt), epithelial–mesenchymal transition and NF-B37 as well as E6 and E7 oncogenes and micro-RNAs (miRNAs) are involved in the HPV-induced cervical cancer.38
Table 1.
Chronic inflammation-related cancers.
Cancer | Associated inflammatory stimuli | References |
---|---|---|
Acute myeloid leukemia | Infection with VGS | 46 |
Bladder cancer | Infection with Schistosoma haematobium | 25 |
Breast cancer | Infection with GBS and obesity | 63 |
Cervical cancer | Infections with HPV or HIV and obesity | 26, 59, 68 |
Cholangiocarcinoma | Clonorchis sinensis and primary sclerosing cholangitis | 25 |
Colorectal cancer | Infections with E. coli, Streptococcus bovis, Fusobacterium nucleatum and Porphyromonas species, inflammatory bowel diseases (ulcerative colitis and Crohn's disease) and obesity | 41, 45, 57, 59, 67 |
Endometrial carcinoma | Barrett's esophagitis and obesity | 59 |
Gall bladder carcinoma | Infection with Salmonella, gall bladder stone-associated chronic cholecystitis and obesity | 42 |
Gastric cancer | Chronic gastric (H pylori) and obesity | 25, 40 |
Haematologic malignancies | Infection with Enterococci faecium | 48 |
Liver cancer | Infections with HBV and HCV, alcohol/non-alcohol-related liver disease and obesity | 27, 31 |
Lung cancer | Infection with Chlamydia pneumoniae, COPD, inflammation caused by asbestos, infections, smoking, obesity and silica | 47, 66, 72 |
Melanoma | UV irradiation-associated skin inflammation | 80 |
Nasopharyngeal carcinoma | Infection with EBV | 28 |
Oral carcinoma | Infections with Fusobacterium nucleatum and Porphyromonas species | 44 |
Pancreatic cancer | Infections with H pylori, Fusobacterium nucleatum and Porphyromonas species, periodontal disease, pancreatitis, obesity | 43 |
Prostate cancer | prostatitis, exposure to Cadmium and pesticides | 24, 78, 79 |
Abbreviations: VSG, viridans group streptococci; GBS, group B streptococcal; HPV, human papilloma virus; HIV, human immunodeficiency virus; HBV, hepatitis B virus; HCV, hepatitis C virus; E. coli, Escherichia coli; COPD, chronic obstructive pulmonary disease; UV, ultraviolet; PM2.5, particulate matter 2.5; EBV, Epstein–Barr virus; H pylori, Helicobacter pylori.
Similar to viruses, several bacterial infections are related to cancer as well. Helicobacter pylori infection is the strongest risk factor for infection-induced cancer worldwide.25 Untreated infection with Helicobacter pylori leads to gastritis, peptic ulcer and eventually gastric cancer through colonization, immune escape and disease induction.39 Although approximately 40%–80% population contracts the viral infection, less than 3% develop into gastric cancer.40 Other examples include Escherichia coli (E. coli) and Streptococcus bovis infections that cause colorectal cancer41; Salmonella infections that cause gall bladder cancer42; Fusobacterium nucleatum and Porphyromonas species that cause colorectal cancer, oral carcinoma and pancreatic cancer43, 44, 45; viridans group streptococci (VGS) infections that cause acute myeloid leukaemia46; Chlamydia pneumoniae infections that cause lung cancer47; Enterococcus faecium infection that causes hematologic malignancies and group B streptococcus (S. GBS) infection that causes breast cancer.48,49
Infections activate multiple cells after being recognized by receptors such as toll-like receptors (TLRs) and nucleotide-binding oligomerization-domain protein-like receptors (NLRs).50 These cells lead to the release of pro-inflammatory cytokines and chemokines such as IL-1, IL-6 and IL-8, which in turn recruit and activate immune cells to produce cytokines, chemokines and free radicals.51,52 These molecules activate host cells through signaling pathways, such as stimulator of interferon genes (STING) and NF-B pathways, and subsequently alter the expression of anti-apoptotic, pro-proliferative and growth factors, thereby promoting cancer progression.53,54
-
2)
Metabolic disorders such as obesity, type 2 diabetes mellitus, insulin resistance and non-alcoholic fatty liver disease are a hallmark of low-grade inflammation, which leads to cancer susceptibilities.55, 56, 57, 58 In 2015, in France, excess weight contributed to 5.4% of cancer cases, including 4500 breast cancer cases, 3400 colon cancer cases, 2500 endometrial cancer cases and 2600 kidney cancer cases.59 In the United States of America, the incidence of obesity-related cancers increased by 7%, whereas the incidence of most other cancers decreased by 13%.60 Metabolic diseases are characterized by abnormal cytokine production (tumor necrosis factor- [TNF-] and IL-6); overexpressed proteins (granulocyte macrophage colony-stimulating factor [GM-CSF] and matrix metalloproteinase [MMP] 9), infiltrated macrophages, neutrophils and eosinophils, which create a pro-tumorigenic environment resulting in accelerated cancer progression and metastasis, stimulation of angiogenesis and reduced patient survival.61, 62, 63, 64, 65
-
3)
Inflammatory bowel disease, sepsis, prostatitis, chronic obstructive pulmonary disease (COPD) and other inflammatory diseases are considered as risk factors for cancer.66, 67, 68, 69, 70 Treatment with non-steroidal anti-inflammatory agents decreases the incidence and mortality of several cancers.4,5 Inflammatory diseases are characterized by the presence of various immune cells, epithelial cells, stromal cells, cytokines and chemokines in the micro-environment, which not only damage tissues but also promote the genetic and epigenetic alterations in normal cells to drive carcinogenesis.71,72 Oncogenes such as Kirsten rat sarcoma 2 viral oncogene homolog (KRAS) and several molecular signaling cascades such as NF-B, Jak family tyrosine kinases (JAK)/STAT, MAPK, Wnt/-catenin and PI3K are activated in those inflammatory diseases, leading to the production of chemokines and adhesion mediators to recruit immune cells, the production of cytokines (IL-1, IL-6, IL-17, IL-21 and TNF-), growth factors (granulocyte colony stimulating factor [G-CSF], GM-CSF and macrophage colony stimulating factor [M-CSF]) and cyclooxygenase (COX) 2 to promote the proliferation and survival of cancer cells and the expression of proteases (MMP 7, MMP 9, MMP 10 and urokinase-type plasminogen activator [uPA]) to promote cancer invasion and metastatic escape.73,74
-
4)
Environmental exposure (chemical, physical and biological stimuli) can cause a loss of homeostasis and promote a pro-neoplastic inflammatory environment.75 Cigarette smoke induced the accumulation of airway macrophages, lymphocytes, neutrophils and monocytes, which causes chronic inflammation, thereby resulting in airway obstruction respiratory symptoms and cancer.76 Inhalable dusts, mineral fibers and particulate matter result in oxidative stress, which initiates the synthesis of pulmonary inflammation mediators in lung epithelial cells and the activation of MAPK and NF-B signaling pathways and eventually induces lung cancer.77 Environmental exposure to cadmium and pesticides increases the risk of prostate cancer.78,79 Ultraviolet (UV) radiation directly affects the skin and leads to DNA damage and reactive oxygen species (ROS) production, followed by the activation of inflammation in the skin tissue, which favors tumorigenesis.80
Acute inflammation in cancer: both the good and evil
In 1868, P. Bruns, a German physician, observed cancer regression in patients with severe streptococcal infection, which provided evidence for an opposite role of inflammation in the development of cancer.81 In 1891, William Coley, an American oncologist, employed this phenomenon and successfully induced cancer regression by injecting extracts of gram-negative bacteria to patients with cancer.82 After more than 50 years, M. Shear was the first to isolate and confirm that the active component of the bacterial extracts was lipopolysaccharide (LPS) and proved its ability to inhibit cancer progression in animals.83 LPS stimulates the immune system by activating TLR4 and releasing TNF-, which not only resist bacteria but also inhibit cancer progression.84 Currently, bacteria- and virus-induced inflammatory response is designed to treat various cancers.85,86 Bacillus Calmette-Guerin (BCG)-induced immune response was associated with cytokine secretion (IL-10/12/18, TNF-, GM-CSF and interferon [IFN]-) and the accumulation of immune cells (NK cells and CD4+ and CD8+ T cells); BCG is widely used for urinary bladder cancer treatment.87 Salmonella typhimurium, Clostridium and other genera were also reported to reduce cancer progression, suppress angiogenesis and metastasis and increase survival in both mouse models and human trials.88,89
However, evidence suggests that acute inflammation promotes cancer progression and metastasis. Standard interventional procedures of cancer diagnosis (biopsies) and treatment (surgeries) or mechanical trauma is sufficient to induce an acute local inflammatory state.90 In such cases, inflammatory cells (macrophages, CD4+ T cells and neutrophils), cytokines, chemokines and growth factors accumulate to form a distant metastatic micro-environment, which may favor cancer progression and invasion.91 Anti-inflammatory treatments (ibuprofen) before and after biopsy significantly reduced the development of lung metastases triggered by the biopsy.92 In addition, once acute inflammation leads to prolonged inflammation, the accompanying cancer rate may increase greatly.
Impact of inflammation with different scopes on cancer
We previously described the influence of different durations of inflammation on cancer. We now describe the relationship between inflammation with different scopes and cancer. According to the scope, inflammation can be divided into systemic inflammation and local inflammation. Inflammation can be induced by some diseases or cancer itself. Currently, it is believed that the prognosis of patients with cancer is determined not only by the characteristics of cancer itself but also by cancer-associated inflammation.93 Systemic inflammation is closely associated with clinical symptoms, which indicate the presence and progression of cancer.94 Cytokines, inflammatory proteins and immune cells are present and easily detectable in systemic circulation.95 Local inflammation is confined to intra-cancer, wherein host cells, cancer cells and immune cells communicate with each other to mediate a local immune response.96 Owing to the different scopes of inflammation, their effects are variable on the subsequent outcome of patients with cancer.
Impact of systemic inflammation on cancer
Cumulating evidence provides a proof that systemic inflammation is characterized by a cancer-promoting immune response and serves as a worse prognostic indicator for patients with cancer.97,98 In patients with cancer, several systemic inflammation markers such as C-reactive protein (CRP) and platelet-to-lymphocyte ratio (PLR) and neutrophil-to-lymphocyte ratio (NLR) are easily detectable and associated with poor survival outcomes or advanced stages in various cancers, including breast, colorectal and lung cancers (Table S1).99, 100, 101, 102 Diseases such as obesity cause systemic low-grade inflammation through multiple mechanisms.103 Overnutrition leads to the rapid expansion of adipose tissues, resulting in hypoxia and endoplasmic reticulum stress.104 Excess production of circulating insulin, leptin, steroid hormones and adiponectin activate mTOR, PI3K and MAPK pathways, thus promoting cancer progression.105 In addition, altered intestinal microbiota activates TLR signaling pathway and immune cells, thereby releasing various cytokines into systemic circulation.106 The resulting sustained inflammatory response may promote the initiation and development of cancer. In patients with cancer, obesity also contributes to an increased risk of recurrence and poor survival outcomes.107,108
The mechanism of cancer-induced systemic inflammation remains unclear; it may attribute to a non-specific response induced by cancer hypoxia/necrosis or oncogene mutations. For instance, mutations of KRAS or tumor protein (TP) 53, may directly induce the transcription of key pro-inflammatory pathways.109,110 Cancer cells and immune cells infiltrated in cancer tissues release various mediators (IL-6, IL-8, vascular endothelial growth factor [VEGF], M-CSF and GM-CSF), which contributed to systemic inflammation by activating the JAK/STAT pathway and result in the expansion of immature myeloid cells and inactivation or tolerance of cytotoxic T cells.111 Neutrophils, myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) accumulate in the circulation and peripheral tissues, inhibit lymphocyte function and form an immunosuppressive macrophage phenotype, thereby forming a positive feedback loop and amplifying the entire process.112 Sustained systemic inflammation further leads to the recruitment and accumulation of immune cells and soluble factors that maintain proliferation, metastasis and neovascularization while suppressing anti-cancer function.113 This is the mechanism by which inflammation and cancer initiate and sustain each other. Excess inflammatory mediators result in alterations in body metabolism and neurological and endocrine functions, leading to cancer cachexia syndromes that causes weight loss, fatigue, anorexia and fevers.114
Impact of local inflammation on cancer
Several studies emphasized that local inflammation, measured based on the density of tumor-infiltrating lymphocytes (TILs), indicated a better prognosis in multiple cancers.115,116 TILs, consisting of a heterogeneous population of lymphocytes such as T cells, B cells, macrophages and NK cells, are localized in the cancer stroma or intra-epithelium.117 The densities of CD3+ and CD8+ TILs exhibited a positive relationship with improved cancer survival, while the effect of TIL-B cells and CD4 TILs was not consistent. Given the positive role of TILs in cancer prognosis, researchers considered mobilizing TILs for cancer treatment. TILs from patients’ blood, lymph nodes or cancer tissues were expanded ex vivo and were adoptively transferred into cancer patients through preparative lymphodepletion and subsequent support of IL-2.118 With this treatment, an approximately 50% response rate was achieved in patients with metastatic melanoma.119 In order to achieve individualized cancer treatments, T cells from peripheral blood were genetically modified for developing T cell receptor-engineered T cell (TCR-T) and chimeric antigen receptor T cell (CAR-T) therapies. Adoptive transfer of modified CD4+ T cells into cancer-bearing mice recruited and activated dendritic cells and subsequently primed CD8+ TILs to exert continuous cytotoxicity to cancers.120
Local treatment-induced inflammation was recently used in combination therapies for cancer. A sub-population of cancer is called ‘cold’ cancer, which exhibits a non-inflamed micro-environment and low response rates to immunotherapy.121 Bacteria/virus-based vaccines (BCG and T-vec) selectively accumulate and replicate in cancer and induce an inflammatory response confined to cancer tissues.122 Combined with chemotherapy, radiotherapy and immunotherapy (cytotoxic T-lymphocyte antigen [CTLA]-4 or programmed cell death protein-1 [PD-1] blockade), these vaccines may induce an inflammatory micro-environment that consists of increased active anticancer T cells and various molecules (IL-2, TNF and IFN), significantly suppress the progression of poorly immunogenic cancer and prolong the overall survival of patients.123,124
However, in some cases, local inflammation may serve as a ‘fertile soil’ to allow cancer progression and metastasis.125 For instance, physical trauma, which evoked local inflammation and wound healing reaction, induced an inflammatory milieu of cytokines, chemokines and growth factors that augmented angiogenesis and favored cancer invasion and evasion of immune surveillance.126,127 Carrageenan-induced acute local inflammation promoted cancer progression by increasing the expression of prostanoids and pro-inflammatory cytokines.128
Impact of inflammation with different sequences on cancer
In addition to the duration and scope of inflammation, the sequence of inflammation is also crucial in cancer development. Chronic inflammation, whether occurring before or after cancer, can promote the occurrence, development and metastasis of cancer. However, the role of inflammation with other forms remains uncertain. The inflammation-then-cancer and reverse cancer-then-inflammation models may lead to opposite effects on cancer progression, and also induced different intra-tumoral immune infiltration.
Sepsis, resulting from acute severe infections, exerts a dysregulated inflammatory reaction in the early stage and an immunosuppressive response in the late stage, which is very similar to immune dysregulation of cancer.129 Recently, some researchers reported that patients who survived bacterial sepsis were more likely to develop malignancies.130, 131, 132 Researchers established a sequential double-hit mice model of polymicrobial sepsis followed by subcutaneous cancer inoculation and found that sepsis promoted cancer development with an increase in cancer-associated granulocytic MDSCs, Tregs or macrophages.133, 134, 135
However, when the sequence was reversed by establishing a cancer-then-sepsis mice model, researchers observed that local and metastatic cancer progression was inhibited by TLR 4-related-enhanced NK cell viability.136 Chen et al. infected cancer-bearing mice with a malaria parasite and found that cancer progression and metastasis were significantly inhibited.137 Furthermore, a clinical study demonstrated that when nine patients with bladder cancer received intra-vesical administration of coxsackievirus 21 (CAVATAK), one patient achieved complete resolution of cancer.138 CAVATAK caused evident inflammatory changes within non-muscle invasive bladder cancer tissues through the upregulation of immune checkpoint-inhibitory genes such as programmed cell death ligand-1 (PD-L1) and lymphocyte-activation gene 3 (LAG3) and induction of the innate activator retinoic acid inducible gene-1 (RIG-1).138 However, when BALB/C mice bearing 4T1 lung metastases were intra-peritoneally injected with 10 mg of LPS, lung weight and pleural lesions of mice were increased, which was accompanied by increased angiogenesis and vascular permeability.139,140 The experimental results suggest that inflammation exerts either a potent pro-cancer function or inhibits cancer progression and metastatic spread, depending on the cancer line and stage and experimental model.
Bacteria/virus-based cancer immunotherapies
BCG, imiquimod and T-vec were approved by the Food and Drug Administration (FDA) for the treatment of bladder cancer, basal cell carcinoma and melanoma, respectively.141,142 Monophosphoryl lipid A (MPLA) was approved by the FDA for use in vaccines for preventing cancers such as cervical cancer.143 Other bacteria and viruses used alone or in combination have entered the clinical trials for cancer treatment (Table 2 and Table 3). As mentioned before, bacterial or viral therapies possess reliable prospects for treating cancer. Bacteria such as Salmonella, Listeria monocytogenes and Clostridium and viruses such as myxoma virus and reovirus, prefer to grow in the hypoxic environment of solid tumors.144,145 Yu et al. found that gram-negative or gram-positive bacteria injected intravenously into mice with syngeneic and xenogeneic cancer as well as spontaneous cancer entered and replicated in the cancer tissue.146 In addition, the clearance of bacteria and virus is inhibited in cancer owing to impaired immunosurveillance or abnormal signaling pathways (IFN, TLR and JAK/STAT).147 Furthermore, besides the direct anti-cancer function, the immune response induced by bacteria and viruses is conducive to cancer elimination.148,149 Oncolytic viruses and C. novyi or Salmonella infections lead to the release of novel tumor antigens and the expression of pathogen-associated and damage-associated molecular patterns, which promote the maturation of dendritic cells and subsequent activation of NK and CD8+ T cell-mediated anti-tumor response.150,151 Moreover, Salmonella infection downregulates the number of Tregs and inhibits angiogenesis to alter the immunosuppressive micro-environment and delay cancer progression.152 In addition, some bacteria and viruses can be used as vectors to carry drugs for enhancing cancer immunotherapy.9 E. coli, engineered to release the nanobody antagonist of CD47 in the tumor site, activates the tumor-infiltrating T cells and results in reliable and systemic anti-tumor immunity and rapid tumor regression.153
Table 2.
Bacteria-based therapies in cancer treatment.
Bacteria-based therapy | Cancer | Status |
---|---|---|
BCG | Bladder cancer | FDA approved |
MPLA | HPV-induced CC | FDA approved |
APS001F | Advanced or metastatic solid tumors | Phase I and II clinical trial (NCT01562626, recruiting) |
C. novyi NT | Solid tumors | Phase I clinical trial (NCT01924689, completed) |
C. novyi NT | Solid tumors | Phase I clinical trial (NCT01118819, terminated) |
C. novyi NT | Tumors | Phase I clinical trial (NCT00358397, terminated) |
Klebsiella pneumoniae (QBKPN SSI) | NSCLC | Phase II clinical trial (NCT02256852, completed) |
L. monocytogenes (ADXS11-001) | CIN | Phase II clinical trial (NCT01116245, terminated) |
L. monocytogenes (ADXS11-001) | CC, recurrent CC | Phase II clinical trial (NCT01266460, active, not recruiting) |
L. monocytogenes (ADXS11-001) | HPV-16 +ve oropharyngeal carcinoma | Phase I clinical trial (NCT01598792, terminated) |
L. monocytogenes (ADXS11-001) | Head and neck cancer, HNSCC, HPV positive oropharyngeal SCC | Phase II clinical trial (NCT02002182, active, not recruiting) |
L. monocytogenes (ADXS11-001) | Metastatic/recurrent and stage IVA, IVB and III CC, cervical adenosquamous cell carcinoma, cervical SCC, cervical small cell carcinoma | Phase I and II clinical trial (NCT02164461, completed) |
L. monocytogenes (ADXS11-001) | Anal and rectal cancer | Phase II clinical trial (NCT02399813, completed) |
L. monocytogenes (ADXS11-001) | High risk and advanced CC | Phase II clinical trial (NCT02853604, active, not recruiting) |
L. monocytogenes (ADU-623) | Astrocytic tumors, GBM, AA, brain tumor | Phase I clinical trial (NCT01967758, completed) |
L. monocytogenes (CRS-100) | Metastatic cancer and liver cancer | Phase I clinical trial (NCT00327652, completed) |
L. monocytogenes (CRS-207) | Malignant epithelial mesothelioma, adenocarcinoma of the pancreas and ovaries, NSCLC | Phase I clinical trial (NCT00585845, terminated) |
L. monocytogenes JNJ-64041757 | NSCLC | Phase I clinical trial (NCT02592967, terminated) |
L. monocytogenes JNJ-64041809 | Castration-resistant prostate neoplasms | Phase I clinical trial (NCT02625857, completed) |
L. monocytogenes pLADD | CRC | Phase I clinical trial (NCT03189030, active, not recruiting) |
S. Typhimurium (VNP20009) | Unspecified adult solid tumor | Phase I clinical trial (NCT0004216, completed) |
S. Typhimurium (VNP20009) | Cancer and metastatic cancer | Phase I clinical trial (NCT0004988, completed) |
S. Typhimurium (VNP20009) | Unspecified adult solid tumor | Phase I clinical trial (NCT0006254, completed) |
S. Typhimurium (VNP20009) | Liver cancer, biliary cancer | Phase I clinical trial (NCT01099631, completed) |
S. Typhimurium (VXM01) | Stage IV pancreatic cancer | Phase I clinical trial (NCT01486329, completed) |
S. Typhimurium (VXM01) | CRC | Phase I clinical trial (NCT02718430, completed) |
S. Typhimurium (VXM01) | Glioblastoma | Phase I clinical trial (NCT02718443, completed) |
Streptococcus pyogenes and Serratia marcescens | Melanoma, sarcoma, GIST, head and neck cancer, TCC, prostate cancer, ovarian carcinoma, esophageal cancer, breast cancer | Phase I clinical trial (NCT00623831, completed) |
DTP, BCG, Measles virus, Serratia marcescens and Streptococcus pneumoniae | Lung cancer | Phase I and II clinical trial (NCT02333474, completed) |
Abbreviations: BCG, Bacillus Calmette-Guerin; MPLA, Monophosphoryl lipid A; HPV, human papillomavirus; CC, cervical cancer; C., Clostridium; L., Listeria; CNS, central nervous system; NSCLC, non-small cell lung cancer; CIN, cervical intraepithelial neoplasia; HNSCC, head and neck squamous cell carcinoma; SCC, squamous cell carcinoma; 5-FU, fluorouracil; MPM, malignant pleural mesothelioma; GAC, gastric adenocarcinoma; EAC, esophageal adenocarcinoma; CRC, colorectal cancer; GBM, glioblastoma multiforme; AA, anaplastic astrocytoma; S., Salmonella; GIST, gastrointestinal stromal tumor; TCC, transitional cell carcinoma; DTP, Diphtheria.
Table 3.
Virus-based therapies in cancer treatment.
Virus-based therapy | Cancer | Status |
---|---|---|
IMQ | Basal cell carcinoma | FDA approved |
T-vec | Melanoma | FDA approved |
Adenovirus | ||
CG0070 | TCC, bladder cancer | Phase I clinical trial (NCT00109655, unknown) |
CG0070 | TCC, bladder cancer, carcinoma in situ with or without papillary tumors | Phase II and III clinical trial (NCT01438112, terminated) |
CG0070 | Bladder cancer | Phase II clinical trial (NCT02365818, completed) |
DNX-2401 | Brain cancer | Phase I clinical trial (NCT00805376, completed) |
DNX-2401 | Glioblastoma or gliosarcoma | Phase I clinical trial (NCT02197169, completed) |
DNX-2401 | Brain glioma | Phase I clinical trial (NCT03178032, completed) |
DNX-2401 | Recurrent high-grade glioma | Phase I clinical trial (NCT03896568, recruiting) |
Enadenotucirev | Solid tumors of epithelial origin, metastatic CRC, metastatic bladder cancer | Phase I and II clinical trial (NCT02028442, completed) |
LOAd703 | Pancreatic cancer | Phase I and II clinical trial (NCT02705196, recruiting) |
LOAd703 | Pancreatic cancer, ovarian cancer, biliary carcinoma, CRC | Phase I and II clinical trial (NCT03225989, recruiting) |
OBP-301 | Hepatocellular carcinoma | Phase I clinical trial (NCT02293850, recruiting) |
OBP-301 | Stage III and IV melanoma | Phase II clinical trial (NCT03190824, active, not recruiting) |
ONCOS-102 | Solid tumors | Phase I clinical trial (NCT01598129, completed) |
VCN-01 | refractory retinoblastoma | Not applicable (NCT03284268, recruiting) |
Coxsackievirus | ||
CAVATAK | Solid tumors | Phase I clinical trial (NCT00636558, completed) |
CAVATAK | Head and neck cancer | Phase I clinical trial (NCT00832559, terminated) |
CAVATAK | Stage IIIc and IV melanoma | Phase II clinical trial (NCT01227551, completed) |
CAVATAK | Melanoma | Phase I clinical trial (NCT01636882, completed) |
CAVATAK | Non-small invasive bladder cancer | Phase I clinical trial (NCT02316171, completed) |
CAVATAK | Advanced/metastatic melanoma | Phase II clinical trial (NCT04152863, recruiting) |
HSV | ||
G207 | Recurrent brain cancer | Phase I and II clinical trial (NCT00028158, completed) |
G207 | Progressive or recurrent supratentorial brain tumor | Phase I clinical trial (NCT02457845, recruiting) |
G207 | Recurrent or refractory cerebellar brain tumors | Phase I clinical trial (NCT03911388, recruiting) |
NV1020 | CRC metastatic to the liver | Phase I and II clinical trial (NCT00149396, completed) |
M032 | Recurrent malignant glioma | Phase I clinical trial (NCT02062827, recruiting) |
HSV1716 | Non-CNS solid tumors | Phase I clinical trial (NCT00931931, completed) |
HSV1716 | Malignant pleural mesothelioma | Phase I and II clinical trial (NCT01721018, completed) |
HF10 | Refractory head and neck cancer or solid tumors with cutaneous and/or superficial lesions | Phase I clinical trial (NCT01017185, completed) |
HF10 | Solid tumors | Phase I clinical trial (NCT02428036, completed) |
rQNestin | Recurrent malignant glioma | Phase I clinical trial (NCT03152318, recruiting) |
C134 | recurrent GBM | Phase I clinical trial (NCT03657576, recruiting) |
RP1 | Melanoma (skin, uveal, ocular), bladder cancer, non-melanoma skin cancer | Phase I and II clinical trial (NCT03767348, recruiting) |
RP1 | Advanced cutaneous SCC | Phase I clinical trial (NCT04349436, not yet recruiting) |
GEN2 | Hepatocellular carcinoma, metastatic cancer | Phase I clinical trial (NCT04313868, recruiting) |
ONCR-177 | Advanced or refractory cutaneous, subcutaneous or metastatic nodal solid tumors | Phase I clinical trial (NCT04348916, recruiting) |
T3011 | Advanced cutaneous or subcutaneous malignancies | Phase I clinical trial (NCT04370587, not yet recruiting) |
MV | ||
MV | Advanced NSCLC | Phase I and II clinical trial (NCT008282022, unknown) |
MV-CEA | Recurrent GBM | Phase I clinical trial (NCT00390299, completed) |
MV-NIS | Ovarian and primary peritoneal cavity cancer | Phase I clinical trial (NCT00408590, completed) |
MV-NIS | Malignant pleural mesothelioma | Phase I clinical trial (NCT01503177, active, not recruiting) |
MV-NIS | Recurrent or metastatic SCC of the head and neck cancer, metastatic breast cancer | Phase I clinical trial (NCT01846091, active, not recruiting) |
MV-NIS | Multiple myeloma | Phase II clinical trial (NCT02192775, completed) |
MV-NIS | Ovarian, fallopian, or peritoneal cancer | Phase II clinical trial (NCT02364713, recruiting) |
MV-NIS | Recurrent malignant peripheral nerve sheath tumor | Phase I clinical trial (NCT02700230, recruiting) |
MV-NIS | Recurrent Medulloblastoma or ATRT | Phase I clinical trial (NCT02962167, recruiting) |
MV-NIS | Bladder cancer | Phase I clinical trial (NCT03171493, recruiting) |
MV mixed vaccine | Lung cancer | Phase I and II clinical trial (NCT02333474, completed) |
Abbreviations: IMQ, Imiquimod; T-vec, Talimogene laherparepvec; TCC, transitional cell carcinoma; CRC, colorectal cancer; HNSCC, head and neck squamous cell carcinoma; HSV, herpes simplex virus; CAVATAK, coxsackievirus 21; CNS, central nervous system; GBM, glioblastoma multiforme; MV, measles virus; MV-CEA, carcinoembryonic antigen-expressing measles virus; MV-NIS, measles virus genetically engineered to produce human thyroidal sodium iodine symporter; MSC, mesenchymal stem cells; NSCLC, non-small cell lung cancer; SCC, squamous cell carcinoma; ATRT, atypical teratoid/rhabdoid tumor.
Conclusions and future perspectives
The effects of inflammation on cancer are significantly different; therefore, different situations should be considered to suppress or promote inflammation for achieving the best response (Fig. 1). Regardless of the form of inflammation, as long as it occurs before the cancer, it should be suppressed. Chronic inflammation is always detrimental. Anti-inflammatory measures like non-steroidal drugs and specialized pro-resolving lipid mediators should be used to prevent the risk and incidence of inflammation-related cancer and avoid the progression from acute inflammation to persistent-chronic inflammation.154 However, when inflammation occurs after cancer formation, factors such as the cause of inflammation and the duration of inflammation should be considered to inhibit or employ inflammation. Surgery- or biopsy-induced wound can stimulate cancer growth via inflammation and angiogenesis, while an acute inflammatory response initiated by recombinant cytokines, TLRs activators and chemotherapeutic agents can effectively promote the infiltration of inflammatory cells into tumor tissues and activate dendritic cell (DC)-mediated anti-tumor T-cell response, slowing the growth of cancer and enhancing the efficacy of anti-cancer therapies.155, 156, 157
Figure 1.
The impact of inflammation on cancer. Inflammation may exert either a potent pro-cancer function or may inhibit cancer progression and metastatic spread, depending on the duration, scope and sequence of inflammation. (A) Several inflammatory conditions promoted cancer progression if inflammation occurred before cancer; (B) Several inflammatory conditions inhibited cancer progression if inflammation occurred after cancer.
Furthermore, although different types of inflammation that cause different outcomes of cancer seem to share similar responses, the involved molecules and immune cells will exert different effects. The increased expression of TNF- in chronic inflammation induces intact TNF/TNF receptor (TNFR) complex and activates the NF-B signaling pathway, which further promote cell survival and tumor growth.158 However, local administration of TNF--caused-acute inflammation induces apoptosis of cancer cells and tumor regression, which is related to the form of modified complex.159,160 Cancer-promoting inflammation selectively recruit various immune cells such as MDSCs, Tregs, tumor-associated fibroblasts and macrophages, which inhibit the CTLs-mediated response, dampen immune surveillance and facilitate the formation of immunosuppressive tumor microenvironments.161 While cancer-inhibiting inflammation induces the M1 polarization of macrophages, promotes the antigen presentation ability of DCs and the infiltration of NK cells, which subsequently active adaptive immunity and eliminate cancer.162,163 Therefore, the essence of inflammation-targeting cancer therapy is how to accurately identify the type of inflammation and selectively promote cancer-inhibiting inflammation and inhibit cancer-promoting inflammation. More studies are also needed to elucidate the interplay between inflammation and cancer.
Author contributions
Writing and original draft preparation: Xinghan Liu; Review & editing: Lijie Yin; Revision and Supervision: Sunan Shen and Yayi Hou. All authors read and agreed to the published version of the manuscript.
Conflict of interests
The authors declare no conflict of interests.
Footnotes
Peer review under responsibility of Chongqing Medical University.
Supplementary data to this article can be found online at https://doi.org/10.1016/j.gendis.2021.09.006.
Contributor Information
Sunan Shen, Email: shensn@nju.edu.cn.
Yayi Hou, Email: yayihou@nju.edu.cn.
Funding
This work was supported by Key Research and Development Program of Jiangsu Province, China (No. BE2019617).
Appendix A. Supplementary data
The following is the Supplementary data to this article:
References
- 1.Sharma A., Schwartz R.A., Swan K.G. Marjolin's warty ulcer. J Surg Oncol. 2011;103(2):193–195. doi: 10.1002/jso.21783. [DOI] [PubMed] [Google Scholar]
- 2.Balkwill F., Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–545. doi: 10.1016/S0140-6736(00)04046-0. [DOI] [PubMed] [Google Scholar]
- 3.Dvorak H.F. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–1659. doi: 10.1056/NEJM198612253152606. [DOI] [PubMed] [Google Scholar]
- 4.Koehne C.H., Dubois R.N. COX-2 inhibition and colorectal cancer. Semin Oncol. 2004;31(2 Suppl 7):12–21. doi: 10.1053/j.seminoncol.2004.03.041. [DOI] [PubMed] [Google Scholar]
- 5.Flossmann E., Rothwell P.M. Effect of aspirin on long-term risk of colorectal cancer: consistent evidence from randomised and observational studies. Lancet. 2007;369(9573):1603–1613. doi: 10.1016/S0140-6736(07)60747-8. [DOI] [PubMed] [Google Scholar]
- 6.Salem M.L., Attia Z.I., Galal S.M. Acute inflammation induces immunomodulatory effects on myeloid cells associated with anti-tumor responses in a tumor mouse model. J Adv Res. 2016;7(2):243–253. doi: 10.1016/j.jare.2015.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Urban-Wojciuk Z., Khan M.M., Oyler B.L., et al. The role of TLRs in anti-cancer immunity and tumor rejection. Front Immunol. 2019;10 doi: 10.3389/fimmu.2019.02388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Surace L., Scheifinger N.A., Gupta A., et al. Radiotherapy supports tumor-specific immunity by acute inflammation. OncoImmunology. 2016;5(1) doi: 10.1080/2162402X.2015.1060391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wood L.M., Paterson Y. Attenuated Listeria monocytogenes: a powerful and versatile vector for the future of tumor immunotherapy. Front Cell Infect Microbiol. 2014;4 doi: 10.3389/fcimb.2014.00051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Salgado R., Denkert C., Demaria S., et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol. 2015;26(2):259–271. doi: 10.1093/annonc/mdu450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Turner N., Wong H.L., Templeton A., et al. Analysis of local chronic inflammatory cell infiltrate combined with systemic inflammation improves prognostication in stage II colon cancer independent of standard clinicopathologic criteria. Int J Cancer. 2016;138(3):671–678. doi: 10.1002/ijc.29805. [DOI] [PubMed] [Google Scholar]
- 12.Wang J., Wang S., Song X., et al. The prognostic value of systemic and local inflammation in patients with laryngeal squamous cell carcinoma. OncoTargets Ther. 2016;9:7177–7185. doi: 10.2147/OTT.S113307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Germolec D.R., Shipkowski K.A., Frawley R.P., et al. Markers of inflammation. Methods Mol Biol. 2018;1803:57–79. doi: 10.1007/978-1-4939-8549-4_5. [DOI] [PubMed] [Google Scholar]
- 14.Feghali C.A., Wright T.M. Cytokines in acute and chronic inflammation. Front Biosci. 1997;2:d12–d26. doi: 10.2741/a171. [DOI] [PubMed] [Google Scholar]
- 15.Liu C.H., Abrams N.D., Carrick D.M., et al. Imaging inflammation and its resolution in health and disease: current status, clinical needs, challenges, and opportunities. Faseb J. 2019;33(12):13085–13097. doi: 10.1096/fj.201902024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liddiard K., Rosas M., Davies L.C., et al. Macrophage heterogeneity and acute inflammation. Eur J Immunol. 2011;41(9):2503–2508. doi: 10.1002/eji.201141743. [DOI] [PubMed] [Google Scholar]
- 17.Davies M.G., Hagen P.O. Systemic inflammatory response syndrome. Br J Surg. 1997;84(7):920–935. doi: 10.1002/bjs.1800840707. [DOI] [PubMed] [Google Scholar]
- 18.Deng T., Lyon C.J., Bergin S., et al. Obesity, inflammation, and cancer. Annu Rev Pathol. 2016;11:421–449. doi: 10.1146/annurev-pathol-012615-044359. [DOI] [PubMed] [Google Scholar]
- 19.Li R., Zhou R., Zhang J. Function of PM2.5 in the pathogenesis of lung cancer and chronic airway inflammatory diseases. Oncol Lett. 2018;15(5):7506–7514. doi: 10.3892/ol.2018.8355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Niccolai E., Boem F., Emmi G., et al. The link "Cancer and autoimmune diseases" in the light of microbiota: evidence of a potential culprit. Immunol Lett. 2020;222:12–28. doi: 10.1016/j.imlet.2020.03.001. [DOI] [PubMed] [Google Scholar]
- 21.Balkwill F.R., Mantovani A. Cancer-related inflammation: common themes and therapeutic opportunities. Semin Cancer Biol. 2012;22(1):33–40. doi: 10.1016/j.semcancer.2011.12.005. [DOI] [PubMed] [Google Scholar]
- 22.Nejsum L.N., Andersen Å.B. Infection and the role in cancer development. APMIS. 2020;128(2) doi: 10.1111/apm.13030. [DOI] [PubMed] [Google Scholar]
- 23.Omabe M., Ezeani M. Infection, inflammation and prostate carcinogenesis. Infect Genet Evol. 2011;11(6):1195–1198. doi: 10.1016/j.meegid.2011.03.002. [DOI] [PubMed] [Google Scholar]
- 24.Liu G.B., Zhao L., Zhang L., et al. Virus, oncolytic virus and human prostate cancer. Curr Cancer Drug Targets. 2017;17(6):522–533. doi: 10.2174/1568009616666161216095308. [DOI] [PubMed] [Google Scholar]
- 25.de Martel C., Georges D., Bray F., et al. Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis. Lancet Glob Health. 2020;8(2):e180–e190. doi: 10.1016/S2214-109X(19)30488-7. [DOI] [PubMed] [Google Scholar]
- 26.Chelimo C., Wouldes T.A., Cameron L.D., et al. Risk factors for and prevention of human papillomaviruses (HPV), genital warts and cervical cancer. J Infect. 2013;66(3):207–217. doi: 10.1016/j.jinf.2012.10.024. [DOI] [PubMed] [Google Scholar]
- 27.Chuang S.C., La Vecchia C., Boffetta P. Liver cancer: descriptive epidemiology and risk factors other than HBV and HCV infection. Cancer Lett. 2009;286(1):9–14. doi: 10.1016/j.canlet.2008.10.040. [DOI] [PubMed] [Google Scholar]
- 28.Zhang J., Jia L., Tsang C.M., et al. EBV infection and glucose metabolism in nasopharyngeal carcinoma. Adv Exp Med Biol. 2017;1018:75–90. doi: 10.1007/978-981-10-5765-6_6. [DOI] [PubMed] [Google Scholar]
- 29.Li L., Wang H. Heterogeneity of liver cancer and personalized therapy. Cancer Lett. 2016;379(2):191–197. doi: 10.1016/j.canlet.2015.07.018. [DOI] [PubMed] [Google Scholar]
- 30.Fu J., Wang H. Precision diagnosis and treatment of liver cancer in China. Cancer Lett. 2018;412:283–288. doi: 10.1016/j.canlet.2017.10.008. [DOI] [PubMed] [Google Scholar]
- 31.Arzumanyan A., Reis H.M., Feitelson M.A. Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat Rev Cancer. 2013;13(2):123–135. doi: 10.1038/nrc3449. [DOI] [PubMed] [Google Scholar]
- 32.Xie Y. Hepatitis B Virus-associated hepatocellular carcinoma. Adv Exp Med Biol. 2017;1018:11–21. doi: 10.1007/978-981-10-5765-6_2. [DOI] [PubMed] [Google Scholar]
- 33.Kew M.C. Hepatitis B virus x protein in the pathogenesis of hepatitis B virus-induced hepatocellular carcinoma. J Gastroenterol Hepatol. 2011;26(Suppl 1):144–152. doi: 10.1111/j.1440-1746.2010.06546.x. [DOI] [PubMed] [Google Scholar]
- 34.Arbyn M., Weiderpass E., Bruni L., et al. Estimates of incidence and mortality of cervical cancer in 2018: a worldwide analysis. Lancet Glob Health. 2020;8(2):e191–e203. doi: 10.1016/S2214-109X(19)30482-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hu Z., Ma D. The precision prevention and therapy of HPV-related cervical cancer: new concepts and clinical implications. Cancer Med. 2018;7(10):5217–5236. doi: 10.1002/cam4.1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gupta S., Kumar P., Das B.C. HPV: molecular pathways and targets. Curr Probl Cancer. 2018;42(2):161–174. doi: 10.1016/j.currproblcancer.2018.03.003. [DOI] [PubMed] [Google Scholar]
- 37.Zhang W., Wu X., Hu L., et al. Overexpression of human papillomavirus type 16 oncoproteins enhances epithelial-mesenchymal transition via STAT3 signaling pathway in non-small cell lung cancer cells. Oncol Res. 2017;25(5):843–852. doi: 10.3727/096504016X14813880882288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wang X., Tang S., Le S.Y., et al. Aberrant expression of oncogenic and tumor-suppressive microRNAs in cervical cancer is required for cancer cell growth. PLoS One. 2008;3(7) doi: 10.1371/journal.pone.0002557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pacifico L., Anania C., Osborn J.F., et al. Consequences of Helicobacter pylori infection in children. World J Gastroenterol. 2010;16(41):5181–5194. doi: 10.3748/wjg.v16.i41.5181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zeyaullah M., AlShahrani A.M., Ahmad I. Association of Helicobacter pylori infection and host cytokine gene polymorphism with gastric cancer. Chin J Gastroenterol Hepatol. 2021;2021 doi: 10.1155/2021/8810620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Koliarakis I., Messaritakis I., Nikolouzakis T.K., et al. Oral bacteria and intestinal dysbiosis in colorectal cancer. Int J Mol Sci. 2019;20(17) doi: 10.3390/ijms20174146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nagaraja V., Eslick G.D. Systematic review with meta-analysis: the relationship between chronic Salmonella typhi carrier status and gall-bladder cancer. Aliment Pharmacol Ther. 2014;39(8):745–750. doi: 10.1111/apt.12655. [DOI] [PubMed] [Google Scholar]
- 43.Michaud D.S. Role of bacterial infections in pancreatic cancer. Carcinogenesis. 2013;34(10):2193–2197. doi: 10.1093/carcin/bgt249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nagy K.N., Sonkodi I., Szöke I., et al. The microflora associated with human oral carcinomas. Oral Oncol. 1998;34(4):304–308. [PubMed] [Google Scholar]
- 45.Shang F.M., Liu H.L. Fusobacterium nucleatum and colorectal cancer: a review. World J Gastrointest Oncol. 2018;10(3):71–81. doi: 10.4251/wjgo.v10.i3.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lewis V., Yanofsky R., Mitchell D., et al. Predictors and outcomes of viridans group streptococcal infections in pediatric acute myeloid leukemia: from the Canadian infections in AML research group. Pediatr Infect Dis J. 2014;33(2):126–129. doi: 10.1097/INF.0000000000000058. [DOI] [PubMed] [Google Scholar]
- 47.Hua-Feng X., Yue-Ming W., Hong L., et al. A meta-analysis of the association between Chlamydia pneumoniae infection and lung cancer risk. Indian J Cancer. 2015;52(Suppl 2):e112–e115. doi: 10.4103/0019-509X.172506. [DOI] [PubMed] [Google Scholar]
- 48.Zhou X., Arends J.P., Span L.F., et al. Algorithm for pre-emptive glycopeptide treatment in patients with haematologic malignancies and an Enterococcus faecium bloodstream infection. Antimicrob Resist Infect Control. 2013;2(1) doi: 10.1186/2047-2994-2-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Shelburne S.A., 3rd, Tarrand J., Rolston K.V. Review of streptococcal bloodstream infections at a comprehensive cancer care center, 2000-2011. J Infect. 2013;66(2):136–146. doi: 10.1016/j.jinf.2012.11.007. [DOI] [PubMed] [Google Scholar]
- 50.Zhou H., Coveney A.P., Wu M., et al. Activation of both TLR and NOD signaling confers host innate immunity-mediated protection against microbial infection. Front Immunol. 2019;9 doi: 10.3389/fimmu.2018.03082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Moss S.F., Blaser M.J. Mechanisms of disease: inflammation and the origins of cancer. Nat Clin Pract Oncol. 2005;2(2):90–97. doi: 10.1038/ncponc0081. [DOI] [PubMed] [Google Scholar]
- 52.Piazuelo M.B., Riechelmann R.P., Wilson K.T., et al. Resolution of gastric cancer-promoting inflammation: a novel strategy for anti-cancer therapy. Curr Top Microbiol Immunol. 2019;421:319–359. doi: 10.1007/978-3-030-15138-6_13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sokolova O., Naumann M. Crosstalk between DNA damage and inflammation in the multiple steps of gastric carcinogenesis. Curr Top Microbiol Immunol. 2019;421:107–137. doi: 10.1007/978-3-030-15138-6_5. [DOI] [PubMed] [Google Scholar]
- 54.He L., Xiao X., Yang X., et al. STING signaling in tumorigenesis and cancer therapy: a friend or foe? Cancer Lett. 2017;402:203–212. doi: 10.1016/j.canlet.2017.05.026. [DOI] [PubMed] [Google Scholar]
- 55.Tilg H., Zmora N., Adolph T.E., et al. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol. 2020;20(1):40–54. doi: 10.1038/s41577-019-0198-4. [DOI] [PubMed] [Google Scholar]
- 56.Pinheiro P.S., Callahan K.E., Jones P.D., et al. Liver cancer: a leading cause of cancer death in the United States and the role of the 1945-1965 birth cohort by ethnicity. JHEP Rep. 2019;1(3):162–169. doi: 10.1016/j.jhepr.2019.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Campisciano G., de Manzini N., Delbue S., et al. The obesity-related gut bacterial and viral dysbiosis can impact the risk of colon cancer development. Microorganisms. 2020;8(3) doi: 10.3390/microorganisms8030431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Colditz G.A., Peterson L.L. Obesity and cancer: evidence, impact, and future directions. Clin Chem. 2018;64(1):154–162. doi: 10.1373/clinchem.2017.277376. [DOI] [PubMed] [Google Scholar]
- 59.Lauby-Secretan B., Dossus L., Marant-Micallef C., et al. [Obesity and cancer] Bull Cancer. 2019;106(7–8):635–646. doi: 10.1016/j.bulcan.2019.04.008. [DOI] [PubMed] [Google Scholar]
- 60.The Lancet The link between cancer and obesity. Lancet. 2017;390(10104) doi: 10.1016/S0140-6736(17)32659-4. [DOI] [PubMed] [Google Scholar]
- 61.Wellen K.E., Hotamisligil G.S. Inflammation, stress, and diabetes. J Clin Invest. 2005;115(5):1111–1119. doi: 10.1172/JCI25102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hotamisligil G.S. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867. doi: 10.1038/nature05485. [DOI] [PubMed] [Google Scholar]
- 63.Iyengar N.M., Zhou X.K., Gucalp A., et al. Systemic correlates of white adipose tissue inflammation in early-stage breast cancer. Clin Cancer Res. 2016;22(9):2283–2289. doi: 10.1158/1078-0432.CCR-15-2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.de la Cruz-Merino L., Chiesa M., Caballero R., et al. Breast cancer immunology and immunotherapy: current status and future perspectives. Int Rev Cell Mol Biol. 2017;331:1–53. doi: 10.1016/bs.ircmb.2016.09.008. [DOI] [PubMed] [Google Scholar]
- 65.Reggiani F., Labanca V., Mancuso P., et al. Adipose progenitor cell secretion of GM-CSF and MMP9 promotes a stromal and immunological microenvironment that supports breast cancer progression. Cancer Res. 2017;77(18):5169–5182. doi: 10.1158/0008-5472.CAN-17-0914. [DOI] [PubMed] [Google Scholar]
- 66.De la Garza M.M., Cumpian A.M., Daliri S., et al. COPD-type lung inflammation promotes K-ras mutant lung cancer through epithelial HIF-1α mediated tumor angiogenesis and proliferation. Oncotarget. 2018;9(68):32972–32983. doi: 10.18632/oncotarget.26030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Huang L.C., Merchea A. Dysplasia and cancer in inflammatory bowel disease. Surg Clin. 2017;97(3):627–639. doi: 10.1016/j.suc.2017.01.010. [DOI] [PubMed] [Google Scholar]
- 68.Zabron A., Edwards R.J., Khan S.A. The challenge of cholangiocarcinoma: dissecting the molecular mechanisms of an insidious cancer. Dis Model Mech. 2013;6(2):281–292. doi: 10.1242/dmm.010561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Villaflor V.M., Allaix M.E., Minsky B., et al. Multidisciplinary approach for patients with esophageal cancer. World J Gastroenterol. 2012;18(46):6737–6746. doi: 10.3748/wjg.v18.i46.6737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sfanos K.S., Yegnasubramanian S., Nelson W.G., et al. The inflammatory microenvironment and microbiome in prostate cancer development. Nat Rev Urol. 2018;15(1):11–24. doi: 10.1038/nrurol.2017.167. [DOI] [PubMed] [Google Scholar]
- 71.Itzkowitz S.H., Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol. 2004;287(1):G7–G17. doi: 10.1152/ajpgi.00079.2004. [DOI] [PubMed] [Google Scholar]
- 72.King P.T. Inflammation in chronic obstructive pulmonary disease and its role in cardiovascular disease and lung cancer. Clin Transl Med. 2015;4(1) doi: 10.1186/s40169-015-0068-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Qu X., Tang Y., Hua S. Immunological approaches towards cancer and inflammation: a cross talk. Front Immunol. 2018;9:e563. doi: 10.3389/fimmu.2018.00563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mantovani A., Allavena P., Sica A., et al. Cancer-related inflammation. Nature. 2008;454(7203):436–444. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
- 75.Archer M., Dogra N., Kyprianou N. Inflammation as a driver of prostate cancer metastasis and therapeutic resistance. Cancers. 2020;12(10) doi: 10.3390/cancers12102984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hamilton G., Rath B. Smoking, inflammation and small cell lung cancer: recent developments. Wien Med Wochenschr. 2015;165(19–20):379–386. doi: 10.1007/s10354-015-0381-6. [DOI] [PubMed] [Google Scholar]
- 77.Ma Q.Y., Huang D.Y., Zhang H.J., et al. Exposure to particulate matter 2.5 (PM2.5) induced macrophage-dependent inflammation, characterized by increased Th1/Th17 cytokine secretion and cytotoxicity. Int Immunopharm. 2017;50:139–145. doi: 10.1016/j.intimp.2017.06.019. [DOI] [PubMed] [Google Scholar]
- 78.Rapisarda V., Miozzi E., Loreto C., et al. Cadmium exposure and prostate cancer: insights, mechanisms and perspectives. Front Biosci (Landmark Ed). 2018;23:1687–1700. doi: 10.2741/4667. [DOI] [PubMed] [Google Scholar]
- 79.Silva J.F., Mattos I.E., Luz L.L., et al. Exposure to pesticides and prostate cancer: systematic review of the literature. Rev Environ Health. 2016;31(3):311–327. doi: 10.1515/reveh-2016-0001. [DOI] [PubMed] [Google Scholar]
- 80.Neagu M., Constantin C., Caruntu C., et al. Inflammation: a key process in skin tumorigenesis. Oncol Lett. 2019;17(5):4068–4084. doi: 10.3892/ol.2018.9735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Korniluk A., Koper O., Kemona H., et al. From inflammation to cancer. Ir J Med Sci. 2017;186(1):57–62. doi: 10.1007/s11845-016-1464-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Coley W.B. II. Contribution to the knowledge of sarcoma. Ann Surg. 1891;14(3):199–220. doi: 10.1097/00000658-189112000-00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Seligman A.M., Shear M.J., Leiter J., et al. Chemical alteration of polysaccharide from Serratia marcescens; tumor-necrotizing polysaccharide tagged with radioactive iodine. J Natl Cancer Inst. 1948;9(1):13–18. [PubMed] [Google Scholar]
- 84.Aggarwal B.B., Sung B. The relationship between inflammation and cancer is analogous to that between fuel and fire. Oncology (Williston Park) 2011;25(5):414–418. [PubMed] [Google Scholar]
- 85.Shetab Boushehri M.A., Lamprecht A. TLR4-based immunotherapeutics in cancer: a review of the achievements and shortcomings. Mol Pharm. 2018;15(11):4777–4800. doi: 10.1021/acs.molpharmaceut.8b00691. [DOI] [PubMed] [Google Scholar]
- 86.Tandon Y., Brodell R.T. Local reactions to imiquimod in the treatment of basal cell carcinoma. Dermatol Online J. 2012;18(9) [PubMed] [Google Scholar]
- 87.Askeland E.J., Newton M.R., O'Donnell M.A., et al. Bladder cancer immunotherapy: BCG and beyond. Adv Urol. 2012;2012 doi: 10.1155/2012/181987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Nallar S.C., Xu D.Q., Kalvakolanu D.V. Bacteria and genetically modified bacteria as cancer therapeutics: current advances and challenges. Cytokine. 2017;89:160–172. doi: 10.1016/j.cyto.2016.01.002. [DOI] [PubMed] [Google Scholar]
- 89.Forbes N.S. Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer. 2010;10(11):785–794. doi: 10.1038/nrc2934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Szalayova G., James T.A., Rincon M. A framework for the role of acute inflammation in tumor progression. Breast Cancer Res Treat. 2015;151(2):235–238. doi: 10.1007/s10549-015-3392-5. [DOI] [PubMed] [Google Scholar]
- 91.Coffey J.C., Wang J.H., Smith M.J., et al. Excisional surgery for cancer cure: therapy at a cost. Lancet Oncol. 2003;4(12):760–768. doi: 10.1016/s1470-2045(03)01282-8. [DOI] [PubMed] [Google Scholar]
- 92.Hobson J., Gummadidala P., Silverstrim B., et al. Acute inflammation induced by the biopsy of mouse mammary tumors promotes the development of metastasis. Breast Cancer Res Treat. 2013;139(2):391–401. doi: 10.1007/s10549-013-2575-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Guthrie G.J., Charles K.A., Roxburgh C.S., et al. The systemic inflammation-based neutrophil-lymphocyte ratio: experience in patients with cancer. Crit Rev Oncol Hematol. 2013;88(1):218–230. doi: 10.1016/j.critrevonc.2013.03.010. [DOI] [PubMed] [Google Scholar]
- 94.Laird B.J., McMillan D.C., Fayers P., et al. The systemic inflammatory response and its relationship to pain and other symptoms in advanced cancer. Oncol. 2013;18(9):1050–1055. doi: 10.1634/theoncologist.2013-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Diakos C.I., Charles K.A., McMillan D.C., et al. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15(11):e493–e503. doi: 10.1016/S1470-2045(14)70263-3. [DOI] [PubMed] [Google Scholar]
- 96.Rumba R., Cipkina S., Cukure F., et al. Systemic and local inflammation in colorectal cancer. Acta Med Litu. 2018;25(4):185–196. doi: 10.6001/actamedica.v25i4.3929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Tu X.P., Qiu Q.H., Chen L.S., et al. Preoperative neutrophil-to-lymphocyte ratio is an independent prognostic marker in patients with laryngeal squamous cell carcinoma. BMC Cancer. 2015;15 doi: 10.1186/s12885-015-1727-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.McMillan D.C. Systemic inflammation, nutritional status and survival in patients with cancer. Curr Opin Clin Nutr Metab Care. 2009;12(3):223–226. doi: 10.1097/MCO.0b013e32832a7902. [DOI] [PubMed] [Google Scholar]
- 99.Li B., Zhou P., Liu Y., et al. Platelet-to-lymphocyte ratio in advanced cancer: review and meta-analysis. Clin Chim Acta. 2018;483:48–56. doi: 10.1016/j.cca.2018.04.023. [DOI] [PubMed] [Google Scholar]
- 100.Liu J., Li S., Zhang S., et al. Systemic immune-inflammation index, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio can predict clinical outcomes in patients with metastatic non-small-cell lung cancer treated with nivolumab. J Clin Lab Anal. 2019;33(8) doi: 10.1002/jcla.22964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Templeton A.J., McNamara M.G., Šeruga B., et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J Natl Cancer Inst. 2014;106(6) doi: 10.1093/jnci/dju124. [DOI] [PubMed] [Google Scholar]
- 102.Shi L., Qin X., Wang H., et al. Elevated neutrophil-to-lymphocyte ratio and monocyte-to-lymphocyte ratio and decreased platelet-to-lymphocyte ratio are associated with poor prognosis in multiple myeloma. Oncotarget. 2017;8(12):18792–18801. doi: 10.18632/oncotarget.13320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Monteiro R., Azevedo I. Chronic inflammation in obesity and the metabolic syndrome. Mediat Inflamm. 2010;2010 doi: 10.1155/2010/289645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.de Heredia F.P., Gómez-Martínez S., Marcos A. Obesity, inflammation and the immune system. Proc Nutr Soc. 2012;71(2):332–338. doi: 10.1017/S0029665112000092. [DOI] [PubMed] [Google Scholar]
- 105.Rawla P., Thandra K.C., Sunkara T. Pancreatic cancer and obesity: epidemiology, mechanism, and preventive strategies. Clin J Gastroenterol. 2019;12(4):285–291. doi: 10.1007/s12328-019-00953-3. [DOI] [PubMed] [Google Scholar]
- 106.Fujita K., Hayashi T., Matsushita M., et al. Obesity, inflammation, and prostate cancer. J Clin Med. 2019;8(2) doi: 10.3390/jcm8020201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Jiralerspong S., Goodwin P.J. Obesity and breast cancer prognosis: evidence, challenges, and opportunities. J Clin Oncol. 2016;34(35):4203–4216. doi: 10.1200/JCO.2016.68.4480. [DOI] [PubMed] [Google Scholar]
- 108.Pischon T., Nimptsch K. Obesity and risk of cancer: an introductory overview. Recent Results Cancer Res. 2016;208:1–15. doi: 10.1007/978-3-319-42542-9_1. [DOI] [PubMed] [Google Scholar]
- 109.Crusz S.M., Balkwill F.R. Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol. 2015;12(10):584–596. doi: 10.1038/nrclinonc.2015.105. [DOI] [PubMed] [Google Scholar]
- 110.West N.R., McCuaig S., Franchini F., et al. Emerging cytokine networks in colorectal cancer. Nat Rev Immunol. 2015;15(10):615–629. doi: 10.1038/nri3896. [DOI] [PubMed] [Google Scholar]
- 111.Tuomisto A.E., Mäkinen M.J., Väyrynen J.P. Systemic inflammation in colorectal cancer: underlying factors, effects, and prognostic significance. World J Gastroenterol. 2019;25(31):4383–4404. doi: 10.3748/wjg.v25.i31.4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Murray P.J., Allen J.E., Biswas S.K., et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41(1):14–20. doi: 10.1016/j.immuni.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Roxburgh C.S., McMillan D.C. Cancer and systemic inflammation: treat the tumour and treat the host. Br J Cancer. 2014;110(6):1409–1412. doi: 10.1038/bjc.2014.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Gabay C., Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 1999;340(6):448–454. doi: 10.1056/NEJM199902113400607. [DOI] [PubMed] [Google Scholar]
- 115.Yu X., Zhang Z., Wang Z., et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in breast cancer: a systematic review and meta-analysis. Clin Transl Oncol. 2016;18(5):497–506. doi: 10.1007/s12094-015-1391-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhao Y., Ge X., He J., et al. The prognostic value of tumor-infiltrating lymphocytes in colorectal cancer differs by anatomical subsite: a systematic review and meta-analysis. World J Surg Oncol. 2019;17(1) doi: 10.1186/s12957-019-1621-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Santoiemma P.P., Powell D.J., Jr. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther. 2015;16(6):807–820. doi: 10.1080/15384047.2015.1040960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rosenberg S.A., Dudley M.E. Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr Opin Immunol. 2009;21(2):233–240. doi: 10.1016/j.coi.2009.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rohaan M.W., Wilgenhof S., Haanen J.B.A.G. Adoptive cellular therapies: the current landscape. Virchows Arch. 2019;474(4):449–461. doi: 10.1007/s00428-018-2484-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Nesbeth Y.C., Martinez D.G., Toraya S., et al. CD4+ T cells elicit host immune responses to MHC class II-negative ovarian cancer through CCL5 secretion and CD40-mediated licensing of dendritic cells. J Immunol. 2010;184(10):5654–5662. doi: 10.4049/jimmunol.0903247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Bonaventura P., Shekarian T., Alcazer V., et al. Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol. 2019;10 doi: 10.3389/fimmu.2019.00168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sørensen M.R., Thomsen A.R. Virus-based immunotherapy of cancer: what do we know and where are we going? APMIS. 2007;115(11):1177–1193. doi: 10.1111/j.1600-0643.2007.00802.x. [DOI] [PubMed] [Google Scholar]
- 123.Ariyan C.E., Brady M.S., Siegelbaum R.H., et al. Robust antitumor responses result from local chemotherapy and CTLA-4 blockade. Cancer Immunol Res. 2018;6(2):189–200. doi: 10.1158/2326-6066.CIR-17-0356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Puzanov I., Milhem M.M., Minor D., et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol. 2016;34(22):2619–2626. doi: 10.1200/JCO.2016.67.1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Jones F.S., Rous P. On the cause of the localization of secondary tumors at points of injury. J Exp Med. 1914;20(4):404–412. doi: 10.1084/jem.20.4.404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Chiang A.C., Massagué J. Molecular basis of metastasis. N Engl J Med. 2008;359(26):2814–2823. doi: 10.1056/NEJMra0805239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Nelson D., Ganss R. Tumor growth or regression: powered by inflammation. J Leukoc Biol. 2006;80(4):685–690. doi: 10.1189/jlb.1105646. [DOI] [PubMed] [Google Scholar]
- 128.Raz A., Levine G., Khomiak Y. Acute local inflammation potentiates tumor growth in mice. Cancer Lett. 2000;148(2):115–120. doi: 10.1016/s0304-3835(99)00329-8. [DOI] [PubMed] [Google Scholar]
- 129.Venet F., Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol. 2018;14(2):121–137. doi: 10.1038/nrneph.2017.165. [DOI] [PubMed] [Google Scholar]
- 130.Kristinsson S.Y., Björkholm M., Hultcrantz M., et al. Chronic immune stimulation might act as a trigger for the development of acute myeloid leukemia or myelodysplastic syndromes. J Clin Oncol. 2011;29(21):2897–2903. doi: 10.1200/JCO.2011.34.8540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Titmarsh G.J., McMullin M.F., McShane C.M., et al. Community-acquired infections and their association with myeloid malignancies. Cancer Epidemiol. 2014;38(1):56–61. doi: 10.1016/j.canep.2013.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Liu Z., Mahale P., Engels E.A. Sepsis and risk of cancer among elderly adults in the United States. Clin Infect Dis. 2019;68(5):717–724. doi: 10.1093/cid/ciy530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Llitjos J.F., Auffray C., Alby-Laurent F., et al. Sepsis-induced expansion of granulocytic myeloid-derived suppressor cells promotes tumour growth through Toll-like receptor 4. J Pathol. 2016;239(4):473–483. doi: 10.1002/path.4744. [DOI] [PubMed] [Google Scholar]
- 134.Cavassani K.A., Carson W.F., 4th, Moreira A.P., et al. The post sepsis-induced expansion and enhanced function of regulatory T cells create an environment to potentiate tumor growth. Blood. 2010;115(22):4403–4411. doi: 10.1182/blood-2009-09-241083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Mota J.M., Leite C.A., Souza L.E., et al. Post-sepsis state induces tumor-associated macrophage accumulation through CXCR4/CXCL12 and favors tumor progression in mice. Cancer Immunol Res. 2016;4(4):312–322. doi: 10.1158/2326-6066.CIR-15-0170. [DOI] [PubMed] [Google Scholar]
- 136.Vigneron C., Mirouse A., Merdji H., et al. Sepsis inhibits tumor growth in mice with cancer through Toll-like receptor 4-associated enhanced natural killer cell activity. OncoImmunology. 2019;8(11) doi: 10.1080/2162402X.2019.1641391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chen L., He Z., Qin L., et al. Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity. PLoS One. 2011;6(9) doi: 10.1371/journal.pone.0024407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Annels N.E., Mansfield D., Arif M., et al. Phase I trial of an ICAM-1-targeted immunotherapeutic-coxsackievirus A21 (CVA21) as an oncolytic agent against non muscle-invasive bladder cancer. Clin Cancer Res. 2019;25(19):5818–5831. doi: 10.1158/1078-0432.CCR-18-4022. [DOI] [PubMed] [Google Scholar]
- 139.Pidgeon G.P., Harmey J.H., Kay E., et al. The role of endotoxin/lipopolysaccharide in surgically induced tumour growth in a murine model of metastatic disease. Br J Cancer. 1999;81(8):1311–1317. doi: 10.1038/sj.bjc.6694369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Harmey J.H., Bucana C.D., Lu W., et al. Lipopolysaccharide-induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int J Cancer. 2002;101(5):415–422. doi: 10.1002/ijc.10632. [DOI] [PubMed] [Google Scholar]
- 141.Chang Y.C., Madkan V., Cook-Norris R., et al. Current and potential uses of imiquimod. South Med J. 2005;98(9):914–920. doi: 10.1097/01.smj.0000176712.01491.98. [DOI] [PubMed] [Google Scholar]
- 142.Johnson D.B., Puzanov I., Kelley M.C. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy. 2015;7(6):611–619. doi: 10.2217/imt.15.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gregg K.A., Harberts E., Gardner F.M., et al. Rationally designed TLR4 ligands for vaccine adjuvant discovery. mBio. 2017;8(3) doi: 10.1128/mBio.00492-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Sedighi M., Zahedi Bialvaei A., Hamblin M.R., et al. Therapeutic bacteria to combat cancer; current advances, challenges, and opportunities. Cancer Med. 2019;8(6):3167–3181. doi: 10.1002/cam4.2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Shi T., Song X., Wang Y., et al. Combining oncolytic viruses with cancer immunotherapy: establishing a new generation of cancer treatment. Front Immunol. 2020;11 doi: 10.3389/fimmu.2020.00683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Yu Y.A., Zhang Q., Szalay A.A. Establishment and characterization of conditions required for tumor colonization by intravenously delivered bacteria. Biotechnol Bioeng. 2008;100(3):567–578. doi: 10.1002/bit.21785. [DOI] [PubMed] [Google Scholar]
- 147.Fend L., Yamazaki T., Remy C., et al. Immune checkpoint blockade, immunogenic chemotherapy or IFN-α blockade boost the local and abscopal effects of oncolytic virotherapy. Cancer Res. 2017;77(15):4146–4157. doi: 10.1158/0008-5472.CAN-16-2165. [DOI] [PubMed] [Google Scholar]
- 148.Guo Y., Chen Y., Liu X., et al. Targeted cancer immunotherapy with genetically engineered oncolytic Salmonella typhimurium. Cancer Lett. 2020;469:102–110. doi: 10.1016/j.canlet.2019.10.033. [DOI] [PubMed] [Google Scholar]
- 149.Osuna-Pérez J., García-Ferreras R., Veiga E. From cellular microbiology to bacteria-based next generations of cancer immunotherapies. Cell Microbiol. 2020;22(4) doi: 10.1111/cmi.13187. [DOI] [PubMed] [Google Scholar]
- 150.Avogadri F., Martinoli C., Petrovska L., et al. Cancer immunotherapy based on killing of Salmonella-infected tumor cells. Cancer Res. 2005;65(9):3920–3927. doi: 10.1158/0008-5472.CAN-04-3002. [DOI] [PubMed] [Google Scholar]
- 151.Gelman A.E., Turka L.A. Autoimmunity heats up. Nat Med. 2003;9(12):1465–1466. doi: 10.1038/nm1203-1465. [DOI] [PubMed] [Google Scholar]
- 152.Liu T., Chopra A.K. An enteric pathogen Salmonella enterica serovar Typhimurium suppresses tumor growth by downregulating CD44high and CD4T regulatory (Treg) cell expression in mice: the critical role of lipopolysaccharide and Braun lipoprotein in modulating tumor growth. Cancer Gene Ther. 2010;17(2):97–108. doi: 10.1038/cgt.2009.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Chowdhury S., Castro S., Coker C., et al. Programmable bacteria induce durable tumor regression and systemic antitumor immunity. Nat Med. 2019;25(7):1057–1063. doi: 10.1038/s41591-019-0498-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Zhang Y., Kong W., Jiang J. Prevention and treatment of cancer targeting chronic inflammation: research progress, potential agents, clinical studies and mechanisms. Sci China Life Sci. 2017;60(6):601–616. doi: 10.1007/s11427-017-9047-4. [DOI] [PubMed] [Google Scholar]
- 155.Panigrahy D., Gartung A., Yang J., et al. Preoperative stimulation of resolution and inflammation blockade eradicates micrometastases. J Clin Invest. 2019;129(7):2964–2979. doi: 10.1172/JCI127282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Lee S.H., Cho S.Y., Yoon Y., et al. Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice. Nat Microbiol. 2021;6(3):277–288. doi: 10.1038/s41564-020-00831-6. [DOI] [PubMed] [Google Scholar]
- 157.Zhao H., Wu L., Yan G., et al. Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther. 2021;6(1) doi: 10.1038/s41392-021-00658-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Lin A., Karin M. NF-kappaB in cancer: a marked target. Semin Cancer Biol. 2003;13(2):107–114. doi: 10.1016/s1044-579x(02)00128-1. [DOI] [PubMed] [Google Scholar]
- 159.Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer. 2009;9(5):361–371. doi: 10.1038/nrc2628. [DOI] [PubMed] [Google Scholar]
- 160.Micheau O., Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003;114(2):181–190. doi: 10.1016/s0092-8674(03)00521-x. [DOI] [PubMed] [Google Scholar]
- 161.Malekghasemi S., Majidi J., Baghbanzadeh A., et al. Tumor-associated macrophages: protumoral macrophages in inflammatory tumor microenvironment. Adv Pharmaceut Bull. 2020;10(4):556–565. doi: 10.34172/apb.2020.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Chrisikos T.T., Zhou Y., Slone N., et al. Molecular regulation of dendritic cell development and function in homeostasis, inflammation, and cancer. Mol Immunol. 2019;110:24–39. doi: 10.1016/j.molimm.2018.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Vacca P., Munari E., Tumino N., et al. Human natural killer cells and other innate lymphoid cells in cancer: friends or foes? Immunol Lett. 2018;201:14–19. doi: 10.1016/j.imlet.2018.11.004. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.