Abstract
The flustering rise in cancer incidence along with treatment anomalies has made cancer the second leading cause of death globally. The total annual economic impact of cancer is pronounced and is increasing. Besides the lack of proper curative therapy, treatment associated adverse effects, drug resistance, and tumor relapse are the instigations behind increased morbidity and mortality. Meanwhile, the survival rate has inclined impressively. In the last few decades, cancer treatment has undergone wide refinements aiming towards cancer prevention, complete tumor regression, subsiding treatment adverse effects, improving patient’s life standard and avoiding tumor relapse. Chemotherapy has been successfully extended towards natural, cheaper and bioactive anti-inflammatory agents manifesting potent anticancer activity. Antibody-based cancer therapy has become well established as a vital and effective strategy for treating hematological malignancies as well as solid tumors. Individualized immunotherapy is becoming the forefront of cancer treatment enabling personalized, precise and patient’s cancer mutanome specific adjustable regimen. The emergence of anti-neoangiogenesis and cancer stem cell targeting techniques have dropped cancer recurrence significantly. Advancements in hyperthermia and photodynamic therapies along with improvements in cancer vaccination have declined death rate and amplified survival rate convincingly.
Keywords: Anticancer therapeutics, cancer targeting, conventional and non-conventional chemotherapy, hyperthermia treatment, photodynamic therapy (PDT), monoclonal antibodies (mAb’s)
Introduction
According to WHO one in every six deaths is caused by cancer [1]. Being the second leading cause of death, cancer is responsible for an estimated 9.6 million deaths worldwide in 2018 [1]. Every four minutes, someone dies from cancer in the UK [2]. Approximately 70% of deaths from cancer occur in middle and low-income countries. The total annual economic impact of cancer is pronounced and is increasing. The estimated economic cost of cancer in 2010 was approximately 1.16 trillion US$ annually. Additionally, a study in 2008 reported that 12.7 million new cases of cancer were identified worldwide, and it was projected that 15-17 million new cases would be identified by 2020 [3]. In accordance, there were 17 million new cases in 2018 and the number is estimated to rise to 27.5 million per year by 2040 [3].
A cancerous cell, after gaining sufficient genetic and epigenetic mutations, replicates at higher rates as compared to the normal cell and establishes a surrounding tumor microenvironment. This microenvironment is crucial for growth, proliferation, and migration of cancer cells [3-6]. The primary technique to fight cancer is believed to be the removal of the cancerous tumor through clinical surgery but with some unwanted side effects. Besides, several other therapies like radiotherapy, immunotherapy, chemotherapy, photodynamic therapy, hyperthermia, non-traditional therapy with natural bioactive materials and tumor vaccination are also in practice, improving and adapting continuously to clinical needs [7,8]. The radio-immunotherapy includes the exploitation of immune proteins as radioactivity carriers or as targeted therapeutics [9,10]. A wide range of chemotherapeutics are used to treat cancer, but they lack obligatory selectivity leading to adverse or side effects [11]. Nanomaterial based drug delivery system is emerging as a promising approach to overcome this problem [12] it involves solid gold nanoparticles [13,14], silver nanoparticles [15], iron oxide nanoparticles [16-18], carbon nanotubes [19,20], quantum dots [21-23], liposomes [24,25], niosomes [26-28] and dendrimers [29-31]. These nano-carriers are of great advantages like the improvement in drug solubility and reduced metabolism, increased circulation time and delivery and comparatively higher accumulations in specific tumors [32,33].
Extensive research is done to explore better prospects or refine the current anti-cancer treatment protocols. Cancer treatment is advancing continuously, the death rate has declined, and the survival rate has been improved convincingly. For instance, the cancer survival rate in the UK has doubled in the last 40 years [2]. In the US, five years the combined survival rate for all childhood cancers has inclined to 81% compared to 62% in 1976 [34]. In the last two decades, cancer treatment has undergone wide refinements aiming towards cancer prevention, complete tumor regression, subsiding treatment adverse effects, improving patient’s life standard and minimizing tumor relapse. The review further describes a brief review of advancements and developments in some major anticancer therapies in the recent past.
Therapeutic strategies
Monoclonal antibodies
Monoclonal antibodies (mAbs), replicates of one type of antibody, that identifies and attaches itself to a specific protein produced by the cells. Depending on the targeted protein, mAbs can work in different ways and can be used to treat different types of cancer [35-37]. Over the past 20 years, antibody-based cancer therapy has become well established and it is now one of the most important and effective strategies for treating patients suffering from hematological malignancies and solid tumors (Table 1) [38-40]. Antibody-based cancer therapy dates back to the original interpretations of antigen expressions by tumor cells through serological techniques in the 1960s [41]. The cell surface antigens expressed by human cancer cells have revealed a broad range of overexpressed targets, mutated or selectively expressed in comparison to normal tissues [42]. The identification of suitable antigens for antibody based therapeutics is a crucial task and can act through interceding modifications in the fundamental functions of antigen or receptor and modulation in the immune system or transportation of a specific drug moiety conjugated to an antibody targeting a particular antigen [43,44]. According to some in vitro and animal studies, mAbs targeting malignant cell surface antigens induce apoptosis through direct transmembrane signaling [45]. The target cancerous cells are also killed by mAbs via complement-mediated cytotoxicity [46] and through inducing antibody dependent cellular toxicity [47]. Bujak et al., with the help of phage display technology generated a human DLK1 specific antibody. Human normal adult tissues and human xeno-grafted tumors frozen sections were used to characterize DLK1 expression using this reagent. Nude mice were injected with radio-iodinated preparations having SIP (F8) and SIP (EB3) with pre-subcutaneously grafted U87 tumors (Figure 1), where neoplastic lesions failed to be detected by IV administration of SIP (EB3) after 24 h. It was observed that the placenta developed a weak expression unlikely in most of the organs where DLK1 was undetectable. It was also found that 8 out of 9 tumor types showed a moderate to strong expression. It was suggested that in the light of limited expression of proteins in normal tissue while abundance in the interstitium of neoplastic lesions, DLK1 might be a good target for antibody-mediated pharmaco-delivery strategies [48].
Table 1.
Different monoclonal antibodies (mAb’s) based anticancer therapeutic approaches and their target antigens [45]
Monoclonal antibodies-based therapeutics | Design | Target antigen |
---|---|---|
Antitumor mAb’s | Modified or unmodified IgG | Tumor specific surface antigens |
Radioimmunotherapy | Unmodified IgG or specific mAb’s fragments | Tumor specific surface antigens that are not shed in the systemic circulation |
Antiangiogenesis | Unmodified IgG | Angiogenic host molecules |
T-cell checkpoint blockade | IgG1 and IgG4 | Anticancer T-cell suppressors |
Antibody and drug conjugate | Drug and IgG linked through a linker for tumor specific drug delivery | Tumor specific surface receptors capable of internalizing mAb and associated drug molecules |
Chimeric antigen receptor T-cell | Modification of T-cells for producing mAb’s specific regions through DNA modification (Gene therapy) | Highly tumor specific surface antigens in non-resistant as well as resistant tumors |
Bispecific antibody | T-cell receptors activating and specific regions of cancer specific mAb’s | Tumor associated specific antigens |
Figure 1.
(A) Showing BIAcore Profiles for SIP antibody and anti DLK1 scFv antibody against human DLK1-Fc recombinant fusion protein (B) SIP (F8) and SIP (EB3) radio-iodinated preparations. The results are shown as injected dose % per tumor tissue gram (% ID/g). Adapted From [49].
Tyrosine kinase which belongs to human EGF receptors (HER) acts as the main target for cancer therapeutics with specificity to HER2 and EGFR because of their in tumor genetic aberrations [49]. The overexpression of HER2 could be correlated with many adverse prognostic features like lower steroid hormone receptor expression, large tumor size, and aneuploidy, etc. [50-52]. Amplification of the HER2 gene is an autonomous adverse prognostic factor [53-56]. Trastuzumab (a recombinant monoclonal antibody against HER2) proved to be of clinical significance in first line chemotherapy against breast cancer by achieving a prolonged disease progression time (median, 7.4 vs. 4.6 months; P<0.001), prolonged response duration (median 9.1 vs. 6.1 months; P<0.001), enhanced objective response rate (50% vs. 32%; P<0.001), lesser death rate at 1 year (22% vs. 33%; P = 0.008), prolonged endurance (median, 25.1 vs. 20.3, 20.3 months; P = 0.046) as well as 20% death risk reduction. Cardiac dysfunction was found to be the most adverse event that occurred in 27% of group given trastuzumab, anthracycline, and cyclophosphamide; 8% of the group with cyclophosphamide and anthracycline; 13% of group given trastuzumab and paclitaxel and 1% of the group delivered with alone paclitaxel. Cardiotoxicity could be covered by proper medical management [57]. Gaborit et al., generated some monoclonal antibodies to HER3 and studied their effect on the degradation of HER3, growth inhibition of cultured cells and also the selection of most potent pancreatic cancer cells inhibitor. It was predicted that in comparison to the mechanism of anti-EGFR antibodies commonly used in the treatment of colorectal cancer, the anti-HER3 antibodies might be able to strongly inhibit tumor growth by intercepting autocrine and stroma-tumor interactions. The anti-HER3 Abs proved to be a better option instead of combing different Abs to enhance the anti-tumor effects of antibodies to NRG receptors [49]. The study proposed to strengthen the opportunity assigning NRGs receptors, HER3 as a powerful motivator in tumor progression as was suggested previously by [58-60]. Very recently some good work is on the way to treat cancer using monoclonal antibodies like phosphatidylserine targeting [61], inhibition of immunosuppressive human regulatory T cell activity by mAbs against GARP/TGF-B1 complexes [62] and anti-CD137 and adoptive T cell therapy [63].
Chemotherapy
Chemotherapy is one of the most widely used strategies for cancer treatment. Chemotherapeutic agents are vital in tumor regression and halting recurrence in more than 100 different known types of cancers [64,65]. They are classified based on their modes of action to antimetabolites, antimicrobial agents, alkylating agents, antimetabolites, platinum complexes and antitumor antibiotics [64,65]. Survival rates for malignancies are increasing with the advent of new modified chemotherapeutic treatments [66]. There are 400,000 malignancy survivors in the US [66]. However, despite enhanced efficacy and improved survival rates, side effects and long-term complications associated with chemotherapy are a major concern for patients, survivors, and clinicians [66,67]. Data suggest that mortality and morbidity due to drug related adverse effects account for an estimated 6.5% of total hospitalizations [68,69]. According to the FDA, 22 drugs used in cancer treatment are associated with more than 25 serious side effects including ulceration, anorexia, vomiting, central and peripheral neurotoxicity, malabsorption, anemia, weight loss, fatigue, chemotherapy induced diarrhea (CID) and constipation (CIC), enhanced risk of sepsis and chemotherapy induced peripheral neuropathy (CIPN). Studies demonstrate that survivors are at 8 folds higher risk of cardiovascular related problems leading to deaths, including coronary artery disease, myocardial infarction, congestive heart failure with cardiomyopathy and cerebrovascular event [66,67,69]. Knowing that almost 70% of deaths from cancer occur in lower- and middle-income countries, the cost associated with chemotherapy is itself a major concern further perpetuating the mater [3]. Scientists have been looking for new and better treatment approaches to combat this challenge by improving tolerance, minimizing the cost of treatment and limiting the sequelae of chemotherapy [66,67]. Convincing reports in the literature, emphasize the use of natural cheaper bioactive compounds as non-traditional chemotherapeutic agents [67,70,71]. Moreover, their synergistic combination with traditional chemotherapeutic agents can limit chemotherapy related adverse effects, potentiate anti-cancer activity, may overcome drug resistance and decrease recurrence [67,70,72]. Some of the reported nutraceuticals include terpenes, mushrooms, flavonoids, curcumin and stilbenes [70]. Plenty of nutraceuticals manifest anti-inflammatory activity and might, not only limit the tumor growth but can also regress the tumor mass [73].
Inflammation and cancer
“Hanahan and Weinberg” described the hallmarks of cancer [74,75]. These hallmarks are the biological capabilities of human cancer cells acquired during the multistep development of tumors. They include evading growth suppressors, sustaining proliferative signaling, enabling replicative immortality, resisting cell death, inducing angiogenesis, initiating invasion and enabling metastasis [74,76]. Further progress enlists genome instability, deregulated metabolism, evading immune system and inflammation as the emerging hallmarks. Cancer cells with all these adverse capabilities ultimately orchestrate tumor microenvironment [74,77].
Inflammatory cells are a vital component of the tumor niche or tumor microenvironment [78]. Inflammation stimulates neoplastic activities involving survival, growth, proliferation, and migration [78,79]. Neoplastic cells have co-opted inflammatory receptors and signaling molecules producing an array of mitogenic chemokines and cytokines [78-82]. These chemoattractants attract inflammatory factors that potentiate the inflammatory process. Some of the players of inflammation include mast cells, granulocytes, lymphocytes, macrophages, fibroblasts, and endothelial cells. In addition to infiltrating inflammatory cells, activated fibroblasts, macrophages and mast cells also secrete cytokines, chemokines, and proteolytic enzymes, initiating neoangiogenesis and lymphangiogenesis [72,73,78,83,84]. Proteases promote the tumor cell escape into the surrounding tissues by disrupting the basement membrane surrounding tumor cells [84,85]. Macrophages being one of the main players release pro-angiogenic factors stimulating neo-angiogenesis and lymph-angiogenesis, immune suppressors avoiding anti-tumor activities of the immune system, tumor cell guiding factors promoting metastasis and growth factors fostering growth and motility of tumor cells [81,82,84,85]. These inflammatory factors potentiate survival, tumor growth, angiogenesis, fibroblast migration, invasion and metastatic spread [71,78,81,82].
Moreover, inflammation can also lead to DNA damage through reactive oxygen species (ROS) and reactive nitrogen species (RNS) induction causing both epigenetic alterations and mutations [86,87]. ROS and RNS are produced locally by various inflammatory mediators, leukocytes, and phagocytes [81,82,86].
Cancers can also arise from sites of chronic irritation, infection, and inflammation [84,85]. Chronic inflammation is related to almost 20% of human cancers [88,89]. Tumors perform as wounds that fail to heal. Chronic inflammation poses the threat of DNA damage becoming a risk factor for cancer [86]. A normal cell subjected to frequent DNA impairment during chronic inflammation may experience carcinogenesis becoming a tumor cell [82,84-86]. Patients with Ulcerative Colitis face a 5 to 7-folds greater risk of developing colon cancer. The risk extends to 20-35% as the UC persists for 35-40 years [88,90,91]. Inflammatory bowel disease (IBD) is also described as a risk factor for colon cancer [92]. Furthermore, studies have demonstrated that long term use of conventional anti-inflammatory agents can limit the cancer incidence [79,83,93-96].
Inflammatory pathways
The mechanism of inflammation involves eicosanoids, the arachidonic acid derived lipidic mediators [97]. Arachidonic acid metabolism pathways include cyclooxygenase (COX) and 5-lipooxygenase (5-LO) pathways. Cyclooxygenase pathway is controlled by the COX enzyme which exists in two isoforms, the constitutive form (COX-1) and the inducible form (COX-2) [73]. COX-1 is involved in mucosal protection, platelet activity maintenance, and renal perfusion. While COX-2 is involved in cell proliferation and inflammation is upregulated by growth factors and cytokines [72,73,98,99]. Prostanoid forming cells orchestrate the breakdown of arachidonic acid into prostaglandins (PGs) and/or thromboxanes (TX) in a cell specific complex fashion producing only the desired product [72,73,100]. Lipoxygenase pathway involves 5-Lipoxygenase (5-LO) enzyme breaking down the arachidonic acid into leukotriene (LT) and lipoxins (LX) [97,101,102]. LTs have remarkable chemotactic activity for recruiting inflammatory components and also release inflammatory mediators promoting inflammation [97,103,104]. LTs also have a strong prognosis in asthma, allergic rhinitis, hypertension, arthritis, psoriasis, and atopic dermatitis, atherosclerosis, chronic obstructive pulmonary disease, liver fibrosis, inflammation, cirrhosis and cancer [101,105-110].
Anti-inflammatory agents in cancer prevention and treatment
Anti-inflammatory agents such as non-steroidal anti-inflammatory drugs (NSAIDs) and anti-leukotrienes play an important role in the prevention and treatment of cancer.
NSAIDs and cancer: The main target of NSAIDs is cyclooxygenase (COX) enzyme [97]. COX inhibitors are classified in three different classes, non-selective COX inhibitors (Ibuprofen, diclofenac, naproxen, and ketoprofen), selective COX-2 inhibitors (Rofecoxib, etoricoxib, lumiracoxib, valdecoxib, celecoxib) and partially selective COX inhibitors (Etodolac, nabumetone, and meloxicam) [95,111-114]. Research has shown the enhanced expression of COX-2 is responsible for inflammatory response and its selective inhibition avoids the unwanted inhibition of COX-1, the isoform involved in housekeeping homeostatic and physiological functions [73,94,95,115,116].
COX-2 inhibitors and cancer: Evidence suggests that long term use of NSAIDs such as aspirin, sulindac, piroxicam, indomethacin, ketoprofen, and ibuprofen counteract the development of colon cancer [95,96,116,117]. It is now well established that selective COX-2 inhibitors not only reduce the incidence of cancer but also regress the established premalignant tumors by stimulating the death of neoplastic cells [71,106,118]. COX-2 inhibition is also effective in limiting angiogenesis and inducing apoptosis [73,119,120]. Not only have these lesser side effects of NSAIDs as compared to traditional chemotherapeutic agents provided a strong rationale for their use in chemotherapy [97,119,121]. Motivated by this, NSAIDs have been used alone as well as in combination with the chemotherapeutic agents by the researchers [71,72,122-124]. However, the synergistic effect depends upon a particular combination of drugs. Such studies have encouraged the clinical use of NSAIDs as potential chemotherapeutic agents [97,122,123]. Table 2 summarizes the current studies in which authors have used the aforementioned rationale to mitigate the cancerous mass aiming for lesser side effects along with enhanced anti-tumor effects.
Table 2.
Reported combinations of COX-2 inhibitor with anticancer drugs and their effects (125-134)
COX-2 inhibitor | Combination with anticancer drug | Effect |
---|---|---|
Celecoxib | Cisplatin | Synergistic effect for lung cancer |
Cyclophosphamide | Additive effect | |
5-FU | Synergistic effect | |
Melphalan | Additive effect | |
Vincristine | None | |
Doxorubicin | Synergistic effect for lung cancer | |
Imatinib | Additive effect | |
Docetaxel | Additive effect | |
Carboplatin | Synergistic effect for lung cancer | |
Sorafenib | Additive effect | |
Parataxol | None | |
Transtuzumab | Additive effect | |
OSU03012 | Antagonistic effect | |
Ifosfamide | Synergistic effect for lung cancer | |
Busulfan | Additive effect | |
Chlormethine | Synergistic effect for lung cancer | |
Melphalan | Additive effect | |
Cis-Platin | Synergistic effect for lung cancer | |
Dichloroacetate (DCA) | Additive effect | |
Mitoxantron | None | |
Mitomycin | Additive effect | |
Bleomycins | Antagonistic effect | |
Aclarubicin | Synergistic effect for lung cancer | |
Rofecoxib | Cisplatin | Synergistic effect for lung cancer |
Texans | Additive effect | |
Mitomycin | None | |
Gemcitabine | Additive effect | |
Leucovorin | Antagonistic effect | |
Docetaxel | Synergistic effect for lung cancer | |
Doxorubicin | Additive effect |
COX-1 vs. COX-2 inhibitors: Nonselective traditional NSAIDs are associated with gastrointestinal side effects [111,112,135]. The risk being highest with indomethacin, ketoprofen, and piroxicam, intermediate with naproxen and lowest with ibuprofen and diclofenac [114,136]. These gastrointestinal side effects are associated with the COX-1 inhibition involved in maintaining normal physiologic functions. Selective COX-2 inhibitors emerged as safe alternatives of non-selective NSAIDs because of their selective COX-2 inhibition property having less gastrointestinal toxicity [114,135]. There is strong evidence that COX-2 expression is upregulated in inflammation as well as in cancer [72,76,122]. COX-2 enzyme has a key role in PGs synthesis and vascular endothelial growth factor production (proangiogenic factor) instigating endothelial cell growth, proliferation, migration and angiogenesis [93,137-139]. Several studies demonstrated that selective COX-2 inhibition can impede cancer development and regress tumor mass highlighting the role of COX-2 in cancer progression [71,93,122,123]. However, selective COX-2 inhibitors were found to increase the cardiovascular risks possibly because of selective COX-2 inhibition disturbing the normal Prostacyclin (PGI2)/Thromboxane balance (TXA2) [113,135,136,140,141]. The disturbed balance leads to irreversible platelet aggregation in blood vessels, vasoconstriction and smooth muscle proliferation promoting thrombotic events. Increased risk of acute myocardial infarction may offset their benefits over non-selective NSAIDs [73,113,141-143].
Recent studies also reported inducible COX-1 isoform in particular cells and constitutive COX-2 isoform of the enzyme in kidney and brain cells implying that the distinction between the two enzymes might not be entirely accurate [73,144,145]. Furthermore, researches have also highlighted the possible role of COX-1 enzyme in angiogenesis regulation suggesting that further clarification is required [95,146].
Anti-leukotrines and cancer: There is accumulating evidence indicating the upregulation of 5-LOX expression and leukotriene receptors in various forms of human cancers, such as colon, breast, head and neck, lung and prostate cancers [101,104,147-149]. Their increased expression is associated with pro-angiogenesis, proliferation of tumor cells and is negatively correlated with patient survival [102,104,148]. Moreover, reports demonstrate that inhibition of 5-LOX enzyme activity can induce apoptosis in neoplastic cells leading to regression of tumor masses [101,147,150]. However, other studies suggest that specific leukotriene receptor blockers instead of 5-LOX inhibitors should be used. This is because specific leukotriene receptor blockers do not inhibit the biosynthesis of lipoxins, that 5-LOX products displaying pro-resolution effects, inhibition of airway hyperresponsiveness and potent anti-inflammatory activities [102,150,151].
Targeted therapy
Escaping reticuloendothelial system (RES)
Various techniques have been used by the researchers to enhance the blood circulation time of drug loaded nanoparticles. Studies reveal that nanoparticles with a lesser diameter (<100 nm) and hydrophilic surfaces are capable of avoiding opsonization, escaping RES and are not taken up by liver, spleen, and lung [152-154]. Various surfactants and polymers e.g. polyvinyl pyrrolidone (PVP) and polyethylene glycol (PEG) have been used successfully to avoid RES, providing stealth properties, decreasing clearance, enhancing the blood circulation time and efficiency of the drug [153-156].
PEG coating (PEGylation) has been used extensively for enhancing blood circulation time [154]. Yet there are evidence that PEGylation is only essential until extravasation. Once the nanoparticle has entered the tissue, PEG coating interferes with nanoparticle-cell interaction and endosomal escape leading to significantly compromised intracellular drug delivery [154]. The downside, being referred to as “PEG dilemma” has been tackled with various alternative strategies. These include the use of alternate polymers (e.g. Polyoxazolines, Poly (amino acids), Polybetaines, N-(2-hydroxypropyl) methacrylamide (HPMA), Polyglycerols and Polysaccharides), conditional removal of PEG effect triggered by cellular cues (pH change, enzymatic stimuli, reductive potential) and external cues (thermal stimuli, ultrasonic stimuli) and biomimetic stealth coating using RBC membrane [157-167].
Targeting through enhanced permeability and retention (EPR) effect
Employing the EPR effect has been a key idea in the passive targeting of tumors over the last two decades [153,168,169]. Defective leaky vasculature in tumors due to neo-angiogenesis coupled with faulty and poor lymphatic drainage allows passive targeting of the cancerous cells [168,170]. Studies have shown that passive targeting enables enhanced penetration of drug carriers into the cancerous mass, increasing the therapeutic effects and reducing the side effects by avoiding off targets [169,170].
Tumor-specific targeting (active targeting)
Malignant cells show the Warburg effect i.e. they are often in hypoxic condition due to significantly enhanced metabolic and growth rate [171-173]. Cancerous cells are distinct from normal cells and can be identified based on several differences from the normal cells, for instance, lower pH, higher temperature, less differentiation, lacking normal physiologic functions and overexpression of specific cell surface receptors including transferrin, folate, hyaluronan, glucose, vascular endothelial growth factor (VEGF) and aldehyde dehydrogenase (ALDH) receptors [171-182]. Researchers have successfully utilized ligands for these receptors as cancer cell targeting agents, reducing the unwanted side effects, minimizing the dose required and increasing the efficiency of therapy. For instance, VEGF, ALDH, N-acetyl-d-glucosamine (NAG), folate, glucose, and transferrin have been used as cancer cell homing agents for active targeting of drug loaded nanocarriers [174,175,177-182].
Cancer stem cells
Recent studies revealed a distinct hierarchy of cancer cells in tumor tissue. Cancer stem cell concept (hypothesis) suggests that not all the tumor cells are equal in cancer [77,183-185]. Only a small pool of tumor cells known as “Cancer stem cells (CSC’s)” have the exclusive ability to initiate cancers [183,186]. This is a subpopulation of “Self-sustaining cells” which are self-renewing, multipotent and can develop heterogeneous tumor mass [77,187]. Sometimes also referred to as “Tumor initiating cells”. “Transit amplifying cells” and “Post mitotic differentiated cells” make the bulk of the tumor. Former are rapidly proliferating cells, and the latter are differentiated cells. Both types of cells originate from tumor initiating cells and have no part in tumor initiation [183,185]. Importantly, recent evidence suggests that tumor initiators or cancer stem cells are very resilient and exquisitely resistant to conventional therapies (Radio and chemotherapy) and tend to be the “drivers” of local recurrence in tumors and metastatic spread [187,188]. It has been postulated that, notably, cancer stem cells can escape from conventional treatment protocols by remaining quiescent for extended periods of time [189]. Moreover, these tumor initiating cells have the potential to get activated, proliferate, differentiate, and lead to the establishment of local recurrences in tumors or distant metastases [77,185,189].
Due to the presence of specific markers on their surface, stem cells can be identified and separated from the bulk cells in a tumor [186,190]. For instance, markers such as CD133, CD44 and ALDH (aldehyde dehydrogenase) have been used successfully to recognize highly tumorigenic cancer stem cells in HNSCC [183,184,187,190]. CD44 is a cell surface glycoprotein functioning as a hyaluronic acid receptor and is involved in cell adhesion and migration [191].
Targeting through neo-angiogenesis and tumor vasculature
The immediate environment around the cancer stem cells is attributed as ‘Tumor microenvironment’ or ‘Tumor niche’ [77,189]. The components of tumor niche include non-epithelial stromal cells, vasculature and inflammatory cells [192]. This microenvironment is crucial for growth, proliferation, and migration of cancer stem cells [193]. Therefore, any disruption in the interaction between cancer stem cells and its supportive niche or any damage to this tumor niche itself can limit the growth and proliferation of cancer stem cells [192,193].
Targeting inflammatory cells and angiogenesis are the two recent approaches being used by the researchers to halt tumor growth [73,111,192,194]. Inflammatory cell targeting has been described earlier in the manuscript. Angiogenesis is the consequence of interactions between various regulatory molecules [151]. These regulatory molecules include angiogenesis stimulators and angiogenesis inhibitors. Angiogenesis in a normal physiological process is self-limited by angio-inhibitory molecules [195-197]. Whereas, the balance between stimulators and inhibitors is disrupted during neo-angiogenesis [151]. This is due to the overproduction of angiogenesis stimulatory factors (VEGF, platelet derived growth factor, basic fibroblast growth factor and matrix metalloproteinases) and diminished expression of angiogenesis inhibitory factors (Thrombospondin-1 and 2, angiostatin, endostatin, tissue inhibitors of matrix metalloproteinases and Interferons α, β and γ) [151,195,197]. Neo-angiogenesis initiated by tumor cells enables the tumor mass to grow in uncontrolled fashion beyond limits [195,196]. Extensive research has been done to develop angiogenesis suppressing strategies [97,194,198]. Researchers have found a strong link between hastily neo-angiogenesis and enhanced COX-2 expression and vice versa [71,72,123]. Studies have also shown that specific vascular targeting agents can not only, successfully halt cancer growth but can also regress the tumor mass [194-198]. In 1999 FDA approved the first anti-angiogenic VEGF targeting drug Avastin® (Bevacizumab) [199]. To date, a lot of new anti-angiogenic drugs have been approved by the FDA for cancer [199]. A brief account has been presented in Table 3.
Table 3.
List of anti-angiogenic drugs approved by the FDA for the treatment of cancer
General | Approved Drugs | Mechanism of action | Indications |
---|---|---|---|
Class | (Brands) | ||
Manufacturer | |||
Monoclonal Antibody Therapies | Bevacizumab (Avastin®) | Monoclonal antibodies directed against VEGF or VEGFR. | Metastatic colorectal cancer (mCRC), non-small cell lung cancer (NSCLC), gastroesophageal junction adenocarcinoma, advanced breast cancer (Europe), glioblastoma, metastatic renal cell cancer (RCC), advanced ovarian cancer (Europe) |
Genentech | |||
Ramucirumab (Cyramza®) | |||
Eli Lilly | |||
Small Molecule Tyrosine Kinase Inhibitors (TKIs) | Axitinib (Inlyta®), Pfizer | Tyrosine kinase or multikinase inhibitor that target in general VEGFR-1, -2, -3, TIE2, PDGFR, and FGFR, KIT, RET, RAF, BRAF, and BRAFV600E. | Advanced renal cell carcinoma |
Cabozantinib (Cometriq®), Exelixis | Advanced metastatic colorectal cancer (mCRC) | ||
Lenvatinib (Lenvima®), Eisai | gastrointestinal stromal tumor (GIST) | ||
Pazopanib (Votrient®) | pancreatic neuroendocrine tumors | ||
GlaxoSmithKline | |||
Regorafenib (Stivarga®), Bayer | |||
Sorafenib (Nexavar®), Bayer, Onyx | |||
Sunitinib (Sutent®), Pfizer | |||
Vandetanib (Caprelsa®), AstraZeneca | |||
Inhibitors of mTOR | Temsirolimus (Torisel®) | Inhibitor of mTOR (mammalian target of rapamycin), part of the PI3 kinase/AKT pathway involved in tumor cell proliferation and angiogenesis. | Advanced renal cell carcinoma, Relapsed or refractory mantle cell lymphoma/Non-Hodgkins Lymphoma (European Union) |
Wyeth | |||
Everolimus (Afinitor®) | |||
Novartis | |||
Fusion Protein | Ziv-Aflibercept (ZALTRAP®) | A recombinant fusion protein consisting of VEGF-binding portions from the extracellular domains of human VEGF receptors 1 and 2 fused to the Fc portion of the human IgG1. | Metastatic colorectal cancer |
Regeneron/Sanofi | |||
Other Antiangiogenic Agents | Interferon alfa (Intron® A and Roferon®), Roche, Schering | The precise mechanisms of action are not fully understood, mixed actions, in general, depending upon the specific drug (e.g. endogenous cytokine, possesses immunomodulatory, anti-inflammatory, and antiangiogenic properties etc.). | Hairy Cell Leukemia, Malignant Melanoma, Follicular Lymphoma, AIDS-Related Kaposi’s Sarcoma |
Lenalidomide (Revlimid®), Celgene | Non-small cell lung cancer (NSCLC) | ||
Thalidomide (Thalomid®, Celgene | metastatic colorectal cancer | ||
TAS-102 (Lonsurf®), Taiho | multiple myeloma | ||
rhEndostatin (Endostar/Endu-available only in China), Simcere | |||
Alitretinoin (Panretin®) | |||
Imiquimod (Aldara®) | |||
Polyphenon E (Veregen®) | |||
Vismodegib (Eviredge, Genentech) | |||
Sonidegib (Odomzo, Novartis) |
Photodynamic therapy (PDT)
Photodynamic therapy is a minimally invasive, dual selective and clinically approved therapeutic procedure for cancer treatment [200,201]. The process involves three individually non-toxic components including photosensitizer (PS), specific wavelength radiations (normally in the visible or near infrared region) and oxygen [202,203]. A combination of these simple components in the specific pattern can exert intricate cytotoxic effects on malignant cells [201]. PDT involves the administration of a photosensitizer into the body followed by tumor irradiation with specific wavelength radiations. The wavelength of the radiations corresponds to the maximum absorption band (λmax) of the photosensitizer. Activated photosensitizer transfers this energy to molecular oxygen in its vicinity culminating in highly reactive singlet oxygen species [202,204,205]. Such species have a half-life of only 1 ms but can exert cytotoxic effects by reacting with vital biomolecules [201]. Depending upon the dose and nature of photosensitizer used, the oxygen concentration in the tumor, light dose and time interval between administration of PS and irradiation, PDT can exert local anti-tumor cytotoxic effects, damage tumor vasculature and produce systemic immunity by inducing acute inflammation [200,206]. Tumor homing ligand grafting enables the targeted PS delivery only to the destination malignant cells or tissue. Irradiation is also spatially directed only to the malignant mass area of the body, making the procedure dual selective [203,205].
Hematoporphyrin (an endogenous porphyrin) was the first PS for PDT, approved by FDA in 1995 [201]. Several less toxic and more sensitive photosensitizers (PSs) have been developed and approved for PDT in the recent past including Porfimer sodium (Photofrin) (HPD) (λmax = 630 nm), 5-aminolevulinic acid (ALA) (λmax = 635 nm), ALA esters (λmax = 635 nm), Temoporfin (Foscan) m-tetrahydroxyphenylchlorin (λmax = 652 nm) and Verteporfin (λmax = 690 nm). Several other photosensitizers are under clinical trials [200,201].
The aging field of PDT has been rejuvenated with the advent of activatable bifunctional photosensitizers [201,203,207]. The diverse type of photosensitizers has been studied within the last two decades. Following irradiation, bifunctional PS can show remarkable fluorescence while participating in intersystem crossing (ISC) and producing reactive singlet oxygen species at the same time [203,207]. Such smarter photosensitizers can serve as theranostics enabling fluorescence imaging as well as photodynamic therapy [203].
Furthermore, the emergence of activatable photosensitizers (aPSs) has augmented the attraction for PDT allowing precise control over the treatment [201,206]. aPSs remain quiescent even under illumination. They get activated selectively in the tumor milieu under specific conditions [208,209]. Such tumor associated stimuli include usually lower pH (6.5 to 7.2) as compared to that in normal tissues (7.4), higher glutathione (GSH) concentration, overexpression of specific enzymes and certain receptors, macrophage targeting and inhibition of self quenching in the tumor niche [208-211]. Not only this, dual responsive aPSs can make PDT even more selective [201,212]. Dual responsive aPS generates maximum singlet oxygen species only when the two tumor associated stimuli are present. One such aPSs system was developed by Lau et al. comprising ferrocene quenchers linked to SiPc core via disulfide and hydrazone linkages. Biothiols and slightly acidic conditions i.e. pH ranging from 4.5 to 6.8 can cleave these linkages respectively. Separation of ferrocene quenchers activates aPSs which then display photo-cytotoxic activity in biothiol rich and slightly acidic tumor microenvironment but not in normal tissues [209].
Such remarkable advancements have improved the efficiency of PDT making it dual selective, bifunctional, dual responsive, minimally invasive and precisely controlled therapy [200-204,212]. Being associated with minimal systemic effects, decreased morbidity, negligible cytotoxicity for normal tissues, lack of acquired or intrinsic resistance, selective cytotoxicity for malignant cells, prolonged patient survival, early stage tumor cure and improved quality of life, PDT has emerged as a vital mainstream anti-cancer therapy [203,205-207,211].
Hyperthermia
Hyperthermia (40-45°C for over 30 min) has been applied clinically to regress tumors for the last 20 years [213,214]. Elevated temperatures (41°-43°C) reduce DNA synthesis and halt respiration in human cells [214]. Further heating to about 45°C results in denaturation of chromosome associated proteins. Hyperthermia can also lead to immune system activation, disruption of the plasma membrane, autophagy and inflammation leading to tumor cell destruction [215-218]. Hyperthermia using computer controlled, sophisticated heating devices and invasive multi-sensor thermometry, has been applied successfully for the treatment of several cancers including malignant melanoma, cervical cancer, soft tissue sarcoma, bladder cancer and recurrent breast cancer [214,219,220].
Cancerous conditions (low pH, hypoxia, altered blood supply and poor nutrition) are associated with resistance development against chemotherapy and radiation therapy. Whereas, the damage due to hyperthermia is more likely and pronounced under these conditions [215]. As hyperthermia affects membrane permeability, it can enhance the drug uptake and improve the efficiency of chemotherapy. Thus, hyperthermia as an adjunct therapy can improve the efficiency of chemotherapy and reduce the resistance development against it [213,215]. Moreover, hyperthermia treatment after radiation therapy impedes cell recovery from impaired mitosis and sub lethal radiation damage by limiting DNA synthesis. Therefore, the combination of precisely localized hyperthermia with chemotherapy and radiotherapy as an adjunct therapy has been suggested [213,215].
Despite the availability of precise, computer controlled and sophisticated techniques providing accurate radiation fields and dosimetry, the treatment is limited due to hot spots and damage to normal tissues [214,215]. Studies suggest a temperature around 43°C for 1 hr. as an optimal thermal dose. Whereas, the average clinical temperatures obtained without affecting normal structures are around (40-41°C) [214,221]. Furthermore, with the present regional radiofrequency, interstitial and superficial techniques, various cancerous sites are unreachable. Temperature inhomogeneity can also lead to unsatisfactory results [213-215].
Current research is focused on dealing with such limitations of heat therapy. A lot of work has been done to develop and modify hyperthermia treatment planning, a valuable tool for improving loco-regional heating [213,222]. It improves understanding of hyperthermia treatment not only as stand-alone but also as an adjunct therapy in cancer treatment by providing information about the realized heat distributions [214,222]. Hyperthermia treatment planning has greatly influenced clinical heating techniques. In the past two decades, various new precise, sophisticated and selective hyperthermia technologies have been developed and optimized for loco-regional heating [213,216,223]. These techniques involve dielectric model generation, electromagnetic field calculation techniques (differential techniques, integral equation methods), thermal modeling (continuum models, discrete vasculature models), treatment optimization techniques (SAR-based optimization, temperature-based optimization) [214,215,217,222,224,225].
Several dedicated sensitive hyperthermia treatment planning software packages have been developed for loco-regional heating providing temperature, electromagnetic and SAR calculations, phase amplitude optimization, thermal modeling, basic and more advanced thermal simulations, tissue segmentation and treatment optimization [213,214,222]. These software packages include HyperPlan, AMC DIVA, SEMCAD X (SPAEG), COMSOL, HFSS and CST STUDIO SUITE [213,214,222,226]. Commercial software packages are improving continuously and adapting to real clinical demands [214].
Cancer vaccination
Cancer immunotherapy and cancer immunoprevention refer to the treatment with vaccines that eliminate existing cancer or prevent the development of new cancer, respectively [227,228]. Immunity is the consequence of a complex interplay between innate (antigen nonspecific) and acquired or adaptive (antigen specific) immune systems. The immunity established by the immune system has the potential to eliminate cancerous cells [229]. Neoplastic cells arise routinely and are destroyed by the immune system. Cancers develop when the protective mechanisms of the immune system fail because of genetic and epigenetic mutations [228,230]. T-lymphocytes, capable of distinguishing between normal and cancerous cells, are the key components of anticancer immunity [229]. Cancer cells, driven by oncogenes or DNA mutations, over-express specific antigens or marker peptides [227,231]. Activated T-cells can reject the tumors by recognizing and destroying major histocompatibility complex (MHC) bound cancer specific epitopes. Stimulation of T-lymphocytes is dependent on antigen presenting cells (APCs) that recognize, bind and present cancer specific epitopes to lymphocytes [229,230]. As cancer cells act as poor APCs, the generation of potent anticancer response depends upon the dendritic cells (DCs) [232]. These are bone marrow derived cells that can identify the cancer antigens and present them to lymphocytes. Extremely efficient, DCs develop a link between innate and adaptive immunity and are designated as “natural adjuvants” or “professional APCs” [229,232].
Cancer immunotherapy
Cancer immunotherapy attempts to harness the natural antitumor response of the immune system through cancer specific antigens and adjuvants [231]. DCs play a vital role in cancer vaccination acting as stimulators of lymphocytes, the key players in anticancer immunity. Different strategies have been adopted to induce effector T-lymphocytes which leads to tumor regression and generation of memory T-cells avoiding tumor relapse [228,229].
In one such approach, autologous epitope specific T-lymphocytes are developed and expanded ex-vivo. Then these lymphocytes are reinfused into the patients inducing anticancer response in-vivo [229].
The vaccination approach involves the administration of cancer specific antigens along with immune system adjuvants eliciting T-lymphocytes in-vivo [228-230]. Depending upon the cancer type to be targeted, vaccines consist of either oncogenes or tumor genes (neoepitopic vaccines), with or without adjuvants, such as dendritic cells. Oncogenes refer to the DNA of oncoviruses that are associated with several cancers including liver and cervical cancers [233]. Such vaccines consist of either DNA of oncoviruses or viral antigens (short viral peptide molecules). For instance, HPV (human papillomavirus) vaccines, HAV (hepatitis A virus) and HBV (hepatitis B virus) vaccines have been developed by the researchers [234,235].
Tumor genes are either shared genes (common to many tumors) or unique genes (Specific to a particular tumor) [233]. Neoepitopic vaccines carry tumor genes and/or the specific antigens overexpressed by cancer cells [231]. Studies demonstrate that short, tumor antigen mRNA of mutated alleles or entire mRNA of cancer cells, loaded in DCs can elicit anticancer immunity [230,236]. The possible mechanism involves the tumor antigen production from tumor mRNA (translation) in the host cell, followed by APC action leading to T-lymphocyte stimulation against tumor antigens [236].
Cancer is the result of genetic and epigenetic mutations that are of diverse nature and cancerous clones vary from patient to patient [237]. This inter and intra-lesional heterogeneity can render the therapeutic vaccines impractical. Moreover, cancer clones keep on changing composition making therapeutic vaccines ineffective with time [238]. Recently researchers have unveiled the possibility of “individualized cancer immunotherapy” [237]. This technique involves the identification of a complete range of patient specific cancer mutations (mutanome) and cancer related epitopes. Then these antigens are produced, multiplied and associated with adjuvants in-vitro, followed by administration into the body [237,238]. The individual specific entire repertoire of tumor specific antigens along with adjuvants can stimulate and/or harness the T-lymphocyte mediated anticancer immune response [232]. Tumor heterogeneity and changes in clonal composition can also be dealt with adjusting the patient specific neoplastic vaccine’s composition over time according to the new or altered mutanome [231,237]. Personalized cancer vaccines target the patient specific genetic aberrations with safety and efficacy, showing the potential to become universally applicable tumor specific agnostic cancer treatment [238].
Cancer immunoprevention
Cancer immunoprevention is an approach to prevent infectious and non-infectious tumors employing the same concept of immunotherapy. It involves the use of antibodies, immunostimulators, and vaccines to kick start the body’s natural immune response [227]. HPV, HAV and HBV vaccines are capable of generating anticancer immune response [234,235]. Still, the technique is not entirely feasible for application in normal individuals due to the general risks associated with vaccination as well as the likelihood of autoimmune disease development [239]. However, it is suggested that the population subgroups with increased cancer risk e.g. individuals with pre-neoplastic lesions or history of hereditary cancer should be considered as possible candidates of non-infectious tumor immunoprevention [227,239].
However, to realize anticancer vaccine therapy, various challenges need to be addressed. Selection of best tumor specific antigens and development of appropriate delivery mechanisms need diligent work. Further, the immune suppressive mechanisms in the cancer tissue can render the immunotherapy ineffectual [229,231]. Cancer immune-evasion strategies involve tumor cell intrinsic (escaping immune recognition and elimination) as well as tumor cell extrinsic mechanisms (immune-suppressive tumor microenvironment creation) [231]. As most of the overexpressed cancer antigens are also expressed by normal cells in a controlled fashion, the risk of developing an autoimmune disease cannot be ruled out and requires keen investigation [227,238,239]. Scientists aim to subside these downsides by employing improved techniques. Some of the recent efforts include vaccine design improvements (e.g. single supramolecular peptide conjugate vaccines) and combining cancer vaccines with other anticancer therapies (e.g. vaccines complementing checkpoint strategies, vaccine-chemotherapy combinations, vaccine and adoptive T-cell therapy) [231,232,237]. With such advancements, cancer vaccination is becoming the forefront of cancer therapy and their future seems bright.
Future prospects
Chemotherapy has been improving continuously and it seems that in the future it will be more effective, specific stem cells and tumor niche targeted involving less toxic and cheaper natural anticancer adjuvants. Commercial software packages for hyperthermia treatment planning are improving continuously and adapting to real clinical demands aiming for realized and precisely controlled loco-regional heating. It is likely that in the future, with the advent of more selective and less toxic activatable photosensitizers, photodynamic therapy will continue to be employed as a stand-alone modality as well as in combination with other anticancer techniques such as chemotherapy. Personalized cancer vaccines will be further improved to deal with alterations in person specific clonal composition and tumor heterogeneity showing the potential to become universally applicable, patient centric, resistance free and tumor specific treatment. Extensive research is being done to develop cancer prevention techniques. Cancer prevention techniques have already been introduced, for instance, prevention through anti-inflammatory agents and cancer immunoprevention vaccines. Refinements in these approaches will most likely make cancer prevention possible in the future.
Conclusion
The total annual economic impact of cancer is huge and is on the rise. Poor early diagnosis, severe adverse effects, high cost and recurrence are the main problems associated with conventional cancer treatment. Extensive research is being done to explore better prospects or refine the current anti-cancer treatment protocols. Successful improvisations in monoclonal antibody development have paved the path for cancer immunopreventive vaccines. Chemotherapeutic agents in combination with natural bioactive anticancer substances and other anticancer therapies demonstrate pronounced anti-neoplastic effects. Anti-inflammatory agents have shown potential as cancer preventing and debulking agents. Targeting inflammatory cells, cancer stem cells and neoangiogenesis are the recent approaches being used by the researchers to halt tumor growth and minimize relapse. Photodynamic therapy has emerged as dual selective, bifunctional, dual responsive, minimally invasive and precisely controlled therapy. Several dedicated sensitive hyperthermia treatment planning software packages have been developed for loco-regional heating providing temperature, electromagnetic and SAR calculations. Personalized cancer vaccines target the patient specific genetic aberrations with safety and efficacy, showing the potential to become universally applicable tumor specific agnostic cancer treatment. Cancer treatment is improving continuously, and the survival rate has inclined remarkably.
Acknowledgements
Authors acknowledge the Higher Education Commission (HEC) of Pakistan for their support.
Disclosure of conflict of interest
None.
References
- 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Cancer Research. Cancer statistics for the UK. Cancer Research UK. 2018 [cited 2020 Feb 2]. pp. 1–4. Available from: http://www.cancerresearchuk.org/health-professional/cancer-statistics.
- 3.López-Gómez M, Malmierca E, de Górgolas M, Casado E. Cancer in developing countries: the next most preventable pandemic. The global problem of cancer. Crit Rev Oncol Hematol. 2013;88:117–22. doi: 10.1016/j.critrevonc.2013.03.011. [DOI] [PubMed] [Google Scholar]
- 4.Gheybi H, Adeli M. Supramolecular anticancer drug delivery systems based on linear-dendritic copolymers. Polym Chem. 2015;6:2580–615. [Google Scholar]
- 5.Diao YY, Li HY, Fu YH, Han M, Hu YL, Jiang HL, Tsutsumi Y, Wei QC, Chen DW, Gao JQ. Doxorubicin-loaded PEG-PCL copolymer micelles enhance cytotoxicity and intracellular accumulation of doxorubicin in adriamycin-resistant tumor cells. Int J Nanomedicine. 2011;6:1955–62. doi: 10.2147/IJN.S23099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Oerlemans C, Bult W, Bos M, Storm G, Nijsen JF, Hennink WE. Polymeric micelles in anticancer therapy: targeting, imaging and triggered release. Pharm Res. 2010;27:2569–89. doi: 10.1007/s11095-010-0233-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Feng SS, Chien S. Chemotherapeutic engineering: application and further development of chemical engineering principles for chemotherapy of cancer and other diseases. Chem Eng Sci. 2003;58:4087–114. [Google Scholar]
- 8.Zeng X, Morgenstern R, Nyström AM. Nanoparticle-directed sub-cellular localization of doxorubicin and the sensitization breast cancer cells by circumventing GST-mediated drug resistance. Biomaterials. 2014;35:1227–39. doi: 10.1016/j.biomaterials.2013.10.042. [DOI] [PubMed] [Google Scholar]
- 9.Larson SM, Carrasquillo JA, Cheung NK, Press OW. Radioimmunotherapy of human tumours. Nat Rev Cancer. 2015;15:347–60. doi: 10.1038/nrc3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Milenic D, Garmestani K, Dadachova E, Chappell L, Albert P, Hill D, Schlom J, Brechbiel M. Radioimmunotherapy of human colon carcinoma xenografts using a 213Bi-labeled domain-deleted humanized monoclonal antibody. Cancer Biother Radiopharm. 2004;19:135–47. doi: 10.1089/108497804323071904. [DOI] [PubMed] [Google Scholar]
- 11.Clementi C, Miller K, Mero A, Satchi-Fainaro R, Pasut G. Dendritic poly (ethylene glycol) bearing paclitaxel and alendronate for targeting bone neoplasms. Mol Pharm. 2011;8:1063–72. doi: 10.1021/mp2001445. [DOI] [PubMed] [Google Scholar]
- 12.Brambilla D, Luciani P, Leroux JC. Breakthrough discoveries in drug delivery technologies: the next 30 years. J Control Release. 2014;190:9–14. doi: 10.1016/j.jconrel.2014.03.056. [DOI] [PubMed] [Google Scholar]
- 13.Popovtzer R, Agrawal A, Kotov NA, Popovtzer A, Balter J, Carey TE, Kopelman R. Targeted gold nanoparticles enable molecular CT imaging of cancer. Nano Lett. 2008;8:4593–6. doi: 10.1021/nl8029114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kang B, Mackey MA, El-Sayed MA. Nuclear targeting of gold nanoparticles in cancer cells induces DNA damage, causing cytokinesis arrest and apoptosis. J Am Chem Soc. 2010;132:1517–9. doi: 10.1021/ja9102698. [DOI] [PubMed] [Google Scholar]
- 15.Ovais M, Khalil AT, Raza A, Khan MA, Ahmad I, Islam NU, Saravanan M, Ubaid MF, Ali M, Shinwari ZK. Green synthesis of silver nanoparticles via plant extracts: beginning a new era in cancer theranostics. Nanomedicine. 2016;12:3157–77. doi: 10.2217/nnm-2016-0279. [DOI] [PubMed] [Google Scholar]
- 16.Kievit FM, Zhang M. Surface engineering of iron oxide nanoparticles for targeted cancer therapy. Acc Chem Res. 2011;44:853–62. doi: 10.1021/ar2000277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dilnawaz F, Singh A, Mohanty C, Sahoo SK. Dual drug loaded superparamagnetic iron oxide nanoparticles for targeted cancer therapy. Biomaterials. 2010;31:3694–706. doi: 10.1016/j.biomaterials.2010.01.057. [DOI] [PubMed] [Google Scholar]
- 18.Rosen JE, Chan L, Shieh DB, Gu FX. Iron oxide nanoparticles for targeted cancer imaging and diagnostics. Nanomedicine. 2012;8:275–90. doi: 10.1016/j.nano.2011.08.017. [DOI] [PubMed] [Google Scholar]
- 19.Robinson JT, Welsher K, Tabakman SM, Sherlock SP, Wang H, Luong R, Dai H. High performance in vivo near-IR (> 1 μm) imaging and photothermal cancer therapy with carbon nanotubes. Nano Res. 2010;3:779–93. doi: 10.1007/s12274-010-0045-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ji S, Liu C, Zhang B, Yang F, Xu J, Long J, Jin C, Fu DL, Ni QX, Yu XJ. Carbon nanotubes in cancer diagnosis and therapy. Biochim Biophys Acta. 2010;1806:29–35. doi: 10.1016/j.bbcan.2010.02.004. [DOI] [PubMed] [Google Scholar]
- 21.Biju V, Mundayoor S, Omkumar RV, Anas A, Ishikawa M. Bioconjugated quantum dots for cancer research: present status, prospects and remaining issues. Biotechnol Adv. 2010;28:199–213. doi: 10.1016/j.biotechadv.2009.11.007. [DOI] [PubMed] [Google Scholar]
- 22.Manzoor K, Johny S, Thomas D, Setua S, Menon D, Nair S. Bio-conjugated luminescent quantum dots of doped ZnS: a cyto-friendly system for targeted cancer imaging. Nanotechnology. 2009;20:65102. doi: 10.1088/0957-4484/20/6/065102. [DOI] [PubMed] [Google Scholar]
- 23.Singh SP. Multifunctional magnetic quantum dots for cancer theranostics. J Biomed Nanotechnol. 2011;7:95–7. doi: 10.1166/jbn.2011.1219. [DOI] [PubMed] [Google Scholar]
- 24.Wu SY, Putral LN, Liang M, Chang HI, Davies NM, McMillan NA. Development of a novel method for formulating stable siRNA-loaded lipid particles for in vivo use. Pharm Res. 2009;26:512–22. doi: 10.1007/s11095-008-9766-1. [DOI] [PubMed] [Google Scholar]
- 25.Koning GA, Eggermont AM, Lindner LH, ten Hagen TL. Hyperthermia and thermosensitive liposomes for improved delivery of chemotherapeutic drugs to solid tumors. Pharm Res. 2010;27:1750–4. doi: 10.1007/s11095-010-0154-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tavano L, Aiello R, Ioele G, Picci N, Muzzalupo R. Niosomes from glucuronic acid-based surfactant as new carriers for cancer therapy: preparation, characterization and biological properties. Colloids Surfaces B Biointerfaces. 2014;118:7–13. doi: 10.1016/j.colsurfb.2014.03.016. [DOI] [PubMed] [Google Scholar]
- 27.Alvi IA, Madan J, Kaushik D, Sardana S, Pandey RS, Ali A. Comparative study of transfersomes, liposomes, and niosomes for topical delivery of 5-fluorouracil to skin cancer cells: preparation, characterization, in-vitro release, and cytotoxicity analysis. Anticancer Drugs. 2011;22:774–82. doi: 10.1097/CAD.0b013e328346c7d6. [DOI] [PubMed] [Google Scholar]
- 28.Puvvada N, Rajput S, Kumar BNP, Mandal M, Pathak A. Exploring the fluorescence switching phenomenon of curcumin encapsulated niosomes: in vitro real time monitoring of curcumin release to cancer cells. RSC Adv. 2013;3:2553–7. [Google Scholar]
- 29.Cheng Y, Zhao L, Li Y, Xu T. Design of biocompatible dendrimers for cancer diagnosis and therapy: current status and future perspectives. Chem Soc Rev. 2011;40:2673–703. doi: 10.1039/c0cs00097c. [DOI] [PubMed] [Google Scholar]
- 30.Wang Y, Guo R, Cao X, Shen M, Shi X. Encapsulation of 2-methoxyestradiol within multifunctional poly (amidoamine) dendrimers for targeted cancer therapy. Biomaterials. 2011;32:3322–9. doi: 10.1016/j.biomaterials.2010.12.060. [DOI] [PubMed] [Google Scholar]
- 31.Mollazade M, Nejati-Koshki K, Akbarzadeh A, Zarghami N, Nasiri M, Jahanban-Esfahlan R, Alibakhshi A. PAMAM dendrimers augment inhibitory effects of curcumin on cancer cell proliferation: possible inhibition of telomerase. Asian Pacific J Cancer Prev. 2013;14:6925–8. doi: 10.7314/apjcp.2013.14.11.6925. [DOI] [PubMed] [Google Scholar]
- 32.Maeda H. Macromolecular therapeutics in cancer treatment: the EPR effect and beyond. J Control Release. 2012;164:138–44. doi: 10.1016/j.jconrel.2012.04.038. [DOI] [PubMed] [Google Scholar]
- 33.Oberoi HS, Laquer FC, Marky LA, Kabanov AV, Bronich TK. Core cross-linked block ionomer micelles as pH-responsive carriers for cis-diamminedichloroplatinum (II) J Control Release. 2011;153:64–72. doi: 10.1016/j.jconrel.2011.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Smith RA, Andrews KS, Brooks D, Fedewa SA, Manassaram-Baptiste D, Saslow D, Brawley OW, Wender RC. Cancer screening in the United States, 2018: a review of current American Cancer Society guidelines and current issues in cancer screening. CA Cancer J Clin. 2018;68:297–316. doi: 10.3322/caac.21446. [DOI] [PubMed] [Google Scholar]
- 35.Scott AM, Allison JP, Wolchok JD. Monoclonal antibodies in cancer therapy. Cancer Immun Arch. 2012;12:14. [PMC free article] [PubMed] [Google Scholar]
- 36.Losanno T, Rossi A, Maione P, Napolitano A, Gridelli C. Anti-EGFR and antiangiogenic monoclonal antibodies in metastatic non-small-cell lung cancer. Expert Opin Biol Ther. 2016;16:747–58. doi: 10.1517/14712598.2016.1163333. [DOI] [PubMed] [Google Scholar]
- 37.Gray MJ, Gong J, Nguyen V, Schuler-Hatch M, Hughes C, Hutchins J, et al. Abstract B27: targeting of phosphatidylserine by monoclonal antibody ch1N11 enhances the antitumor activity of immune checkpoint inhibitor PD-1/PD-L1 therapy in orthotopic murine breast cancer models. AACR. 2016 [Google Scholar]
- 38.Diaz LA Jr, Coughlin CM, Weil SC, Fishel J, Gounder MM, Lawrence S, Azad N, O’Shannessy DJ, Grasso L, Wustner J, Ebel W, Carvajal RD. A first-in-human phase I study of MORAb-004, a monoclonal antibody to endosialin in patients with advanced solid tumors. Clin Cancer Res. 2015;21:1281–8. doi: 10.1158/1078-0432.CCR-14-1829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Asundi J, Crocker L, Tremayne J, Chang P, Sakanaka C, Tanguay J, Spencer S, Chalasani S, Luis E, Gascoigne K, Desai R, Raja R, Friedman BA, Haverty PM, Polakis P, Firestein R. An antibody-drug conjugate directed against lymphocyte antigen 6 complex, locus E (LY6E) provides robust tumor killing in a wide range of solid tumor malignancies. Clin Cancer Res. 2015;21:3252–62. doi: 10.1158/1078-0432.CCR-15-0156. [DOI] [PubMed] [Google Scholar]
- 40.Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, Beeram M, Drengler R, Chen C, Smith L, Espino G, Gergich K, Delgado L, Daud A, Lindia JA, Li XN, Pierce RH, Yearley JH, Wu D, Laterza O, Lehnert M, Iannone R, Tolcher AW. Phase I study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin cancer Res. 2015;21:4286–93. doi: 10.1158/1078-0432.CCR-14-2607. [DOI] [PubMed] [Google Scholar]
- 41.Rettig WJ, Old LJ. Immunogenetics of human cell surface differentiation. Annu Rev Immunol. 1989;7:481–511. doi: 10.1146/annurev.iy.07.040189.002405. [DOI] [PubMed] [Google Scholar]
- 42.Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer. 2012;12:278–87. doi: 10.1038/nrc3236. [DOI] [PubMed] [Google Scholar]
- 43.Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol. 2010;10:317–27. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hughes B. Antibody-drug conjugates for cancer: poised to deliver? Nat Rev Drug Discov. 2010;9:665–7. doi: 10.1038/nrd3270. [DOI] [PubMed] [Google Scholar]
- 45.Weiner GJ. Building better monoclonal antibody-based therapeutics. Nat Rev Cancer. 2015;15:361–70. doi: 10.1038/nrc3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Taylor RP. Of mice and mechanisms: identifying the role of complement in monoclonal antibody-based immunotherapy. Haematologica. 2006;91 146a. [PubMed] [Google Scholar]
- 47.Clynes R, Takechi Y, Moroi Y, Houghton A, Ravetch JV. Fc receptors are required in passive and active immunity to melanoma. Proc Natl Acad Sci. 1998;95:652–6. doi: 10.1073/pnas.95.2.652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bujak E, Ritz D, Neri D. A monoclonal antibody to human DLK1 reveals differential expression in Cancer and absence in healthy tissues. Antibodies. 2015;4:71–87. [Google Scholar]
- 49.Gaborit N, Abdul-Hai A, Mancini M, Lindzen M, Lavi S, Leitner O, Mounier L, Chentouf M, Dunoyer S, Ghosh M, Larbouret C, Chardès T, Bazin H, Pèlegrin A, Sela M, Yarden Y. Examination of HER3 targeting in cancer using monoclonal antibodies. Proc Natl Acad Sci U S A. 2015;112:839–44. doi: 10.1073/pnas.1423645112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Narayan M, Wilken JA, Harris LN, Baron AT, Kimbler KD, Maihle NJ. Trastuzumab-induced HER reprogramming in “resistant” breast carcinoma cells. Cancer Res. 2009;69:2191–4. doi: 10.1158/0008-5472.CAN-08-1056. [DOI] [PubMed] [Google Scholar]
- 51.Ménard S, Fortis S, Castiglioni F, Agresti R, Balsari A. HER2 as a prognostic factor in breast cancer. Oncology. 2001;61(Suppl 2):67–72. doi: 10.1159/000055404. [DOI] [PubMed] [Google Scholar]
- 52.Konecny G, Pauletti G, Pegram M, Untch M, Dandekar S, Aguilar Z, Wilson C, Rong HM, Bauerfeind I, Felber M, Wang HJ, Beryt M, Seshadri R, Hepp H, Slamon DJ. Quantitative association between HER-2/neu and steroid hormone receptors in hormone receptor-positive primary breast cancer. J Natl Cancer Inst. 2003;95:142–53. doi: 10.1093/jnci/95.2.142. [DOI] [PubMed] [Google Scholar]
- 53.Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235:177–82. doi: 10.1126/science.3798106. [DOI] [PubMed] [Google Scholar]
- 54.Ross JS, Fletcher JA. The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Stem Cells. 1998;16:413–28. doi: 10.1002/stem.160413. [DOI] [PubMed] [Google Scholar]
- 55.Ross JS, Fletcher JA, Bloom KJ, Linette GP, Stec J, Symmans WF, Pusztai L, Hortobagyi GN. Targeted therapy in breast cancer: the HER-2/neu gene and protein. Mol Cell Proteomics. 2004;3:379–98. doi: 10.1074/mcp.R400001-MCP200. [DOI] [PubMed] [Google Scholar]
- 56.Nielsen DL, Andersson M, Kamby C. HER2-targeted therapy in breast cancer. Monoclonal antibodies and tyrosine kinase inhibitors. Cancer Treat Rev. 2009;35:121–36. doi: 10.1016/j.ctrv.2008.09.003. [DOI] [PubMed] [Google Scholar]
- 57.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
- 58.Khambata-Ford S, Garrett CR, Meropol NJ, Basik M, Harbison CT, Wu S, Wong TW, Huang X, Takimoto CH, Godwin AK, Tan BR, Krishnamurthi SS, Burris HA 3rd, Poplin EA, Hidalgo M, Baselga J, Clark EA, Mauro DJ. Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J. Clin. Oncol. 2007;25:3230–7. doi: 10.1200/JCO.2006.10.5437. [DOI] [PubMed] [Google Scholar]
- 59.Jacobs B, De Roock W, Piessevaux H, Van Oirbeek R, Biesmans B, De Schutter J, Fieuws S, Vandesompele J, Peeters M, Van Laethem JL, Humblet Y, Pénault-Llorca F, De Hertogh G, Laurent-Puig P, Van Cutsem E, Tejpar S. Amphiregulin and epiregulin mRNA expression in primary tumors predicts outcome in metastatic colorectal cancer treated with cetuximab. J. Clin. Oncol. 2009;27:5068–74. doi: 10.1200/JCO.2008.21.3744. [DOI] [PubMed] [Google Scholar]
- 60.Prenen H, De Schutter J, Jacobs B, De Roock W, Biesmans B, Claes B, Lambrechts D, Van Cutsem E, Tejpar S. PIK3CA mutations are not a major determinant of resistance to the epidermal growth factor receptor inhibitor cetuximab in metastatic colorectal cancer. Clin Cancer Res. 2009;15:3184–8. doi: 10.1158/1078-0432.CCR-08-2961. [DOI] [PubMed] [Google Scholar]
- 61.Gray M, Gong J, Nguyen V, Hutchins J, Freimark B. Abstract P4-04-03: targeting of phosphatidylserine by monoclonal antibodies augments the activity of immune checkpoint inhibitor PD-1/PD-L1 therapy in murine breast tumors. AACR. 2016 doi: 10.1186/s13058-016-0708-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Liénart S, Stockis J, Dedobbeleer O, Lucas S. Targeting immunosuppression by Tregs with monoclonal antibodies against GARP. Oncoimmunology. 2016;5:e1074379. doi: 10.1080/2162402X.2015.1074379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Weigelin B, Bolaños E, Rodriguez-Ruiz ME, Martinez-Forero I, Friedl P, Melero I. Anti-CD137 monoclonal antibodies and adoptive T cell therapy: a perfect marriage? Cancer Immunol Immunother. 2016;65:493–7. doi: 10.1007/s00262-016-1818-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2:751–60. doi: 10.1038/nnano.2007.387. [DOI] [PubMed] [Google Scholar]
- 65.Platt VM, Szoka FC Jr. Anticancer therapeutics: targeting macromolecules and nanocarriers to hyaluronan or CD44, a hyaluronan receptor. Mol Pharm. 2008;5:474–86. doi: 10.1021/mp800024g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Loar RW, Noel CV, Tunuguntla H, Colquitt JL, Pignatelli RH. State of the art review: Chemotherapy-induced cardiotoxicity in children. Congenit Heart Dis. 2018;13:5–15. doi: 10.1111/chd.12564. [DOI] [PubMed] [Google Scholar]
- 67.Nurgali K, Jagoe RT, Abalo R. Adverse effects of cancer chemotherapy: anything new to improve tolerance and reduce sequelae? Front Pharmacol. 2018;9:245. doi: 10.3389/fphar.2018.00245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA. 1998;279:1200–5. doi: 10.1001/jama.279.15.1200. [DOI] [PubMed] [Google Scholar]
- 69.Hassett MJ, O’Malley AJ, Pakes JR, Newhouse JP, Earle CC. Frequency and cost of chemotherapy-related serious adverse effects in a population sample of women with breast cancer. J Natl Cancer Inst. 2006;98:1108–17. doi: 10.1093/jnci/djj305. [DOI] [PubMed] [Google Scholar]
- 70.Zhou W, Yan Y, Mi J, Zhang H, Lu L, Luo Q, Li X, Zeng X, Cao Y. Simulated digestion and fermentation in vitro by human gut microbiota of polysaccharides from bee collected pollen of Chinese wolfberry. J Agric Food Chem. 2018;66:898–907. doi: 10.1021/acs.jafc.7b05546. [DOI] [PubMed] [Google Scholar]
- 71.Toomey DP, Murphy JF, Conlon KC. COX-2, VEGF and tumour angiogenesis. Surgeon. 2009;7:174–80. doi: 10.1016/s1479-666x(09)80042-5. [DOI] [PubMed] [Google Scholar]
- 72.Gately S, Li WW Seminars in Oncology. Multiple roles of COX-2 in tumor angiogenesis: a target for antiangiogenic therapy. Elsevier; 2004. pp. 2–11. [DOI] [PubMed] [Google Scholar]
- 73.Claria J, Romano M. Pharmacological intervention of cyclooxygenase-2 and 5-lipoxygenase pathways. Impact on inflammation and cancer. Curr Pharm Des. 2005;11:3431–47. doi: 10.2174/138161205774370753. [DOI] [PubMed] [Google Scholar]
- 74.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 75.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 76.Greenhough A, Smartt HJ, Moore AE, Roberts HR, Williams AC, Paraskeva C, Kaidi A. The COX-2/PGE 2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis. 2009;30:377–86. doi: 10.1093/carcin/bgp014. [DOI] [PubMed] [Google Scholar]
- 77.Krishnamurthy S, Nör JE. Head and neck cancer stem cells. J Dent Res. 2012;91:334–40. doi: 10.1177/0022034511423393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Crusz SM, Balkwill FR. Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol. 2015;12:584–86. doi: 10.1038/nrclinonc.2015.105. [DOI] [PubMed] [Google Scholar]
- 79.Frölich JC. A classification of NSAIDs according to the relative inhibition of cyclooxygenase isoenzymes. Trends Pharmacol Sci. 1997;18:30–4. doi: 10.1016/s0165-6147(96)01017-6. [DOI] [PubMed] [Google Scholar]
- 80.Ungefroren H, Sebens S, Seidl D, Lehnert H, Hass R. Interaction of tumor cells with the microenvironment. Cell Commun Signal. 2011;9:18. doi: 10.1186/1478-811X-9-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Chiba T, Marusawa H, Ushijima T. Inflammation-associated cancer development in digestive organs: mechanisms and roles for genetic and epigenetic modulation. Gastroenterology. 2012;143:550–63. doi: 10.1053/j.gastro.2012.07.009. [DOI] [PubMed] [Google Scholar]
- 82.Ding N, Maiuri AR, O’Hagan HM. The emerging role of epigenetic modifiers in repair of DNA damage associated with chronic inflammatory diseases. Mutat Res Mutat Res. 2019;780:69–81. doi: 10.1016/j.mrrev.2017.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rothwell PM, Price JF, Fowkes FG, Zanchetti A, Roncaglioni MC, Tognoni G, Lee R, Belch JF, Wilson M, Mehta Z, Meade TW. Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials. Lancet. 2012;379:1602–12. doi: 10.1016/S0140-6736(11)61720-0. [DOI] [PubMed] [Google Scholar]
- 84.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Shacter E, Weitzman SA. Chronic inflammation and cancer. Oncology (Williston Park) 2002;16:217–26. [PubMed] [Google Scholar]
- 86.Dizdaroglu M. Oxidatively induced DNA damage: mechanisms, repair and disease. Cancer Lett. 2012;327:26–47. doi: 10.1016/j.canlet.2012.01.016. [DOI] [PubMed] [Google Scholar]
- 87.Kawanishi S, Ohnishi S, Ma N, Hiraku Y, Oikawa S, Murata M. Nitrative and oxidative DNA damage in infection-related carcinogenesis in relation to cancer stem cells. Genes Environ. 2016;38:26. doi: 10.1186/s41021-016-0055-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001;48:526–35. doi: 10.1136/gut.48.4.526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
- 90.Ekbom A, Helmick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer: a population-based study. N Engl J Med. 1990;323:1228–33. doi: 10.1056/NEJM199011013231802. [DOI] [PubMed] [Google Scholar]
- 91.Eaden J, Abrams K, Ekbom A, Jackson E, Mayberry J. Colorectal cancer prevention in ulcerative colitis: a case-control study. Aliment Pharmacol Ther. 2000;14:145–53. doi: 10.1046/j.1365-2036.2000.00698.x. [DOI] [PubMed] [Google Scholar]
- 92.Sano H, Kawahito Y, Wilder RL, Hashiramoto A, Mukai S, Asai K, Kimura S, Kato H, Kondo M, Hla T. Expression of cyclooxygenase-1 and-2 in human colorectal cancer. Cancer Res. 1995;55:3785–9. [PubMed] [Google Scholar]
- 93.Wang D, DuBois RN. The role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene. 2010;29:781–8. doi: 10.1038/onc.2009.421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sheng H, Shao J, Kirkland SC, Isakson P, Coffey RJ, Morrow J, Beauchamp RD, DuBois RN. Inhibition of human colon cancer cell growth by selective inhibition of cyclooxygenase-2. J Clin Invest. 1997;99:2254–9. doi: 10.1172/JCI119400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Thun MJ, Henley SJ, Patrono C. Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, pharmacologic, and clinical issues. J Natl Cancer Inst. 2002;94:252–66. doi: 10.1093/jnci/94.4.252. [DOI] [PubMed] [Google Scholar]
- 96.Chan AT, Ogino S, Fuchs CS. Aspirin and the risk of colorectal cancer in relation to the expression of COX-2. N Engl J Med. 2007;356:2131–42. doi: 10.1056/NEJMoa067208. [DOI] [PubMed] [Google Scholar]
- 97.Goossens L, Pommery N, Pierre Henichart J. COX-2/5-LOX dual acting anti-inflammatory drugs in cancer chemotherapy. Curr Top Med Chem. 2007;7:283–96. doi: 10.2174/156802607779941369. [DOI] [PubMed] [Google Scholar]
- 98.Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR. TIS10, a phorbol ester tumor promoter-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthase/cyclooxygenase homologue. J Biol Chem. 1991;266:12866–72. [PubMed] [Google Scholar]
- 99.O’Banion MK, Sadowski HB, Winn V, Young DA. A serum-and glucocorticoid-regulated 4-kilobase mRNA encodes a cyclooxygenase-related protein. J Biol Chem. 1991;266:23261–7. [PubMed] [Google Scholar]
- 100.Tanioka T, Nakatani Y, Semmyo N, Murakami M, Kudo I. Molecular identification of cytosolic prostaglandin E2 synthase that is functionally coupled with cyclooxygenase-1 in immediate prostaglandin E2 biosynthesis. J Biol Chem. 2000;275:32775–82. doi: 10.1074/jbc.M003504200. [DOI] [PubMed] [Google Scholar]
- 101.Yoshimura R, Matsuyama M, Mitsuhashi M, Takemoto Y, Tsuchida K, Kawahito Y, Sano H, Nakatani T. Relationship between lipoxygenase and human testicular cancer. Int J Mol Med. 2004;13:389–93. [PubMed] [Google Scholar]
- 102.Nathan RA, Kemp JP Antileukotriene Working Group. Efficacy of antileukotriene agents in asthma management. Ann Allergy Asthma Immunol. 2001;86(Suppl 1):24–30. doi: 10.1016/s1081-1206(10)62306-x. [DOI] [PubMed] [Google Scholar]
- 103.Shureiqi I, Wu Y, Chen D, Yang XL, Guan B, Morris JS, Yang P, Newman RA, Broaddus R, Hamilton SR, Lynch P, Levin B, Fischer SM, Lippman SM. The critical role of 15-lipoxygenase-1 in colorectal epithelial cell terminal differentiation and tumorigenesis. Cancer Res. 2005;65:11486–92. doi: 10.1158/0008-5472.CAN-05-2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Barker HE, Cox TR, Erler JT. The rationale for targeting the LOX family in cancer. Nat Rev Cancer. 2012;12:540–52. doi: 10.1038/nrc3319. [DOI] [PubMed] [Google Scholar]
- 105.Creticos PS, Peters SP, Adkinson NF Jr, Naclerio RM, Hayes EC, Norman PS, Lichtenstein LM. Peptide leukotriene release after antigen challenge in patients sensitive to ragweed. N Engl J Med. 1984;310:1626–30. doi: 10.1056/NEJM198406213102502. [DOI] [PubMed] [Google Scholar]
- 106.Celotti F, Durand T. The metabolic effects of inhibitors of 5-lipoxygenase and of cyclooxygenase 1 and 2 are an advancement in the efficacy and safety of anti-inflammatory therapy. Prostaglandins Other Lipid Mediat. 2003;71:147–62. doi: 10.1016/s1098-8823(03)00039-x. [DOI] [PubMed] [Google Scholar]
- 107.Stanke-Labesque F, Hardy G, Caron F, Cracowski J, Bessard G. Inhibition of leukotriene synthesis with MK-886 prevents a rise in blood pressure and reduces noradrenaline-evoked contraction in L-NAME-treated rats. Br J Pharmacol. 2003;140:186–94. doi: 10.1038/sj.bjp.0705405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Mehrabian M, Allayee H, Wong J, Shih W, Wang XP, Shaposhnik Z, Funk CD, Lusis AJ. Identification of 5-lipoxygenase as a major gene contributing to atherosclerosis susceptibility in mice. Circ Res. 2002;91:120–6. doi: 10.1161/01.res.0000028008.99774.7f. [DOI] [PubMed] [Google Scholar]
- 109.Huber M, Kästner S, Schölmerich J, Gerok W, Keppler D. Analysis of cysteinyl leukotrienes in human urine: enhanced excretion in patients with liver cirrhosis and hepatorenal syndrome. Eur J Clin Invest. 1989;19:53–60. doi: 10.1111/j.1365-2362.1989.tb00195.x. [DOI] [PubMed] [Google Scholar]
- 110.Shindo K, Hirai Y, Fukumura M, Koide K. Plasma levels of leukotriene E4 during clinical course of chronic obstructive pulmonary disease. Prostaglandins Leukot Essent Fatty Acids. 1997;56:213–7. doi: 10.1016/s0952-3278(97)90537-8. [DOI] [PubMed] [Google Scholar]
- 111.Day RO, Graham GG. The vascular effects of COX-2 selective inhibitors. 2004 [Google Scholar]
- 112.Bleumink GS, Feenstra J, Sturkenboom MC, Stricker BH. Nonsteroidal anti-inflammatory drugs and heart failure. Drugs. 2003;63:525–34. doi: 10.2165/00003495-200363060-00001. [DOI] [PubMed] [Google Scholar]
- 113.Mason RP, Walter MF, McNulty HP, Lockwood SF, Byun J, Day CA, Jacob RF. Rofecoxib increases susceptibility of human LDL and membrane lipids to oxidative damage: a mechanism of cardiotoxicity. J Cardiovasc Pharmacol. 2006;47:S7–14. doi: 10.1097/00005344-200605001-00003. [DOI] [PubMed] [Google Scholar]
- 114.Conaghan PG. A turbulent decade for NSAIDs: update on current concepts of classification, epidemiology, comparative efficacy, and toxicity. Rheumatol Int. 2012;32:1491–502. doi: 10.1007/s00296-011-2263-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, Wakabayashi N, Saunders B, Shen Y, Fujimura T, Su LK, Levin B, Godio L, Patterson S, Rodriguez-Bigas MA, Jester SL, King KL, Schumacher M, Abbruzzese J, DuBois RN, Hittelman WN, Zimmerman S, Sherman JW, Kelloff G. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med. 2000;342:1946–52. doi: 10.1056/NEJM200006293422603. [DOI] [PubMed] [Google Scholar]
- 116.Rocca B, Secchiero P, Ciabattoni G, Ranelletti FO, Catani L, Guidotti L, Melloni E, Maggiano N, Zauli G, Patrono C. Cyclooxygenase-2 expression is induced during human megakaryopoiesis and characterizes newly formed platelets. Proc Natl Acad Sci U S A. 2002;99:7634–9. doi: 10.1073/pnas.112202999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Piazza GA, Rahm AK, Finn TS, Fryer BH, Li H, Stoumen AL, Pamukcu R, Ahnen DJ. Apoptosis primarily accounts for the growth-inhibitory properties of sulindac metabolites and involves a mechanism that is independent of cyclooxygenase inhibition, cell cycle arrest, and p53 induction. Cancer Res. 1997;57:2452–9. [PubMed] [Google Scholar]
- 118.Elder DJ, Halton DE, Hague A, Paraskeva C. Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug: independence from COX-2 protein expression. Clin Cancer Res. 1997;3:1679–83. [PubMed] [Google Scholar]
- 119.Kashfi K, Rayyan Y, Qiao LL, Williams JL, Chen J, Del Soldato P, Traganos F, Rigas B, Ryann Y. Nitric oxide-donating nonsteroidal anti-inflammatory drugs inhibit the growth of various cultured human cancer cells: evidence of a tissue type-independent effect. J Pharmacol Exp Ther. 2002;303:1273–82. doi: 10.1124/jpet.102.042754. [DOI] [PubMed] [Google Scholar]
- 120.Kawamori T, Rao CV, Seibert K, Reddy BS. Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis. Cancer Res. 1998;58:409–12. [PubMed] [Google Scholar]
- 121.Yeh RK, Chen J, Williams JL, Baluch M, Hundley TR, Rosenbaum RE, Kalala S, Traganos F, Benardini F, del Soldato P, Kashfi K, Rigas B. NO-donating nonsteroidal antiinflammatory drugs (NSAIDs) inhibit colon cancer cell growth more potently than traditional NSAIDs: a general pharmacological property? Biochem Pharmacol. 2004;67:2197–205. doi: 10.1016/j.bcp.2004.02.027. [DOI] [PubMed] [Google Scholar]
- 122.Ghosh N, Chaki R, Mandal V, Mandal SC. COX-2 as a target for cancer chemotherapy. Pharmacol Rep. 2010;62:233–44. doi: 10.1016/s1734-1140(10)70262-0. [DOI] [PubMed] [Google Scholar]
- 123.Bakhle YS. COX-2 and cancer: a new approach to an old problem. Br J Pharmacol. 2001;134:1137–50. doi: 10.1038/sj.bjp.0704365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Hohenforst-Schmidt W, Domvri K, Zogas N, Zarogoulidis P, Petanidis S, Kioseoglou E, Zachariadis G, Kakolyris S, Porpodis K, Gaga M, Huang HD, Kontakiotis T, Zarogoulidis K. COX-2 inhibitors, a potential synergistic effect with antineoplastic drugs in lung cancer. Oncomedicine. 2017;2:28–36. [Google Scholar]
- 125.Arunasree KM, Roy KR, Anilkumar K, Aparna A, Reddy GV, Reddanna P. Imatinib-resistant K562 cells are more sensitive to celecoxib, a selective COX-2 inhibitor: role of COX-2 and MDR-1. Leuk Res. 2008;32:855–64. doi: 10.1016/j.leukres.2007.11.007. [DOI] [PubMed] [Google Scholar]
- 126.Becerra CR, Frenkel EP, Ashfaq R, Gaynor RB. Increased toxicity and lack of efficacy of Rofecoxib in combination with chemotherapy for treatment of metastatic colorectal cancer: a phase II study. Int J cancer. 2003;105:868–72. doi: 10.1002/ijc.11164. [DOI] [PubMed] [Google Scholar]
- 127.Chen M, Yu L, Gu C, Zhong D, Wu S, Liu S. Celecoxib antagonizes the cytotoxic effect of cisplatin in human gastric cancer cells by decreasing intracellular cisplatin accumulation. Cancer Lett. 2013;329:189–96. doi: 10.1016/j.canlet.2012.10.030. [DOI] [PubMed] [Google Scholar]
- 128.Groen HJ, Sietsma H, Vincent A, Hochstenbag MM, van Putten JW, van den Berg A, Dalesio O, Biesma B, Smit HJ, Termeer A, Hiltermann TJ, van den Borne BE, Schramel FM. Randomized, placebo-controlled phase III study of docetaxel plus carboplatin with celecoxib and cyclooxygenase-2 expression as a biomarker for patients with advanced non-small-cell lung cancer: the NVALT-4 study. J. Clin. Oncol. 2011;29:4320–6. doi: 10.1200/JCO.2011.35.5214. [DOI] [PubMed] [Google Scholar]
- 129.Hiľovská L, Jendželovský R, Fedoročko P. Potency of non-steroidal anti-inflammatory drugs in chemotherapy. Mol Clin Oncol. 2015;3:3–12. doi: 10.3892/mco.2014.446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Ponthan F, Wickström M, Gleissman H, Fuskevåg OM, Segerström L, Sveinbjörnsson B, Redfern CP, Eksborg S, Kogner P, Johnsen JI. Celecoxib prevents neuroblastoma tumor development and potentiates the effect of chemotherapeutic drugs in vitro and in vivo. Clin Cancer Res. 2007;13:1036–44. doi: 10.1158/1078-0432.CCR-06-1908. [DOI] [PubMed] [Google Scholar]
- 131.Sugiura T, Saikawa Y, Kubota T, Suganuma K, Otani Y, Watanabe M, Kumai K, Kitajima M. Combination chemotherapy with JTE-522, a novel selective cyclooxygenase-2 inhibitor, and cisplatin against gastric cancer cell lines in vitro and in vivo. In Vivo (Brooklyn) 2003;17:229–33. [PubMed] [Google Scholar]
- 132.Yan YX, Li WZ, Huang YQ, Liao WX. The COX-2 inhibitor Celecoxib enhances the sensitivity of KB/VCR oral cancer cell lines to Vincristine by down-regulating P-glycoprotein expression and function. Prostaglandins Other Lipid Mediat. 2012;97:29–35. doi: 10.1016/j.prostaglandins.2011.07.007. [DOI] [PubMed] [Google Scholar]
- 133.Ye CG, Wu WK, Yeung JH, Li HT, Li ZJ, Wong CC, Ren SX, Zhang L, Fung KP, Cho CH. Indomethacin and SC236 enhance the cytotoxicity of doxorubicin in human hepatocellular carcinoma cells via inhibiting P-glycoprotein and MRP1 expression. Cancer Lett. 2011;304:90–6. doi: 10.1016/j.canlet.2011.01.025. [DOI] [PubMed] [Google Scholar]
- 134.Zatelli MC, Luchin A, Tagliati F, Leoni S, Piccin D, Bondanelli M, Rossi R, degli Uberti EC. Cyclooxygenase-2 inhibitors prevent the development of chemoresistance phenotype in a breast cancer cell line by inhibiting glycoprotein p-170 expression. Endocr Relat Cancer. 2007;14:102938. doi: 10.1677/ERC-07-0114. [DOI] [PubMed] [Google Scholar]
- 135.Dajani EZ, Islam K. Cardiovascular and gastrointestinal toxicity of selective cyclo-oxygenase-2 inhibitors in man. J Physiol Pharmacol. 2008;59(Suppl 2):117–33. [PubMed] [Google Scholar]
- 136.Ng SC, Chan FK. NSAID-induced gastrointestinal and cardiovascular injury. Curr Opin Gastroenterol. 2010;26:611–7. doi: 10.1097/MOG.0b013e32833e91eb. [DOI] [PubMed] [Google Scholar]
- 137.Grösch S, Tegeder I, Niederberger E, Brä utigam L, Geisslinger G. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB J. 2001;15:2742–4. doi: 10.1096/fj.01-0299fje. [DOI] [PubMed] [Google Scholar]
- 138.Ristimäki A, Sivula A, Lundin J, Lundin M, Salminen T, Haglund C, Salminen T, Haglund C, Joensuu H, Isola J. Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Res. 2002;62:632–5. [PubMed] [Google Scholar]
- 139.Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ 3rd. Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res. 1999;59:987–90. [PubMed] [Google Scholar]
- 140.Hernández-Díaz S, Varas-Lorenzo C, García Rodríguez LA. Non-steroidal antiinflammatory drugs and the risk of acute myocardial infarction. Basic Clin Pharmacol Toxicol. 2006;98:266–74. doi: 10.1111/j.1742-7843.2006.pto_302.x. [DOI] [PubMed] [Google Scholar]
- 141.Barnabe C, Martin B, Ghali WA. Systematic review and meta-analysis: anti-tumor necrosis factor α therapy and cardiovascular events in rheumatoid arthritis. Arthritis Care Res (Hoboken) 2011;63:522–9. doi: 10.1002/acr.20371. [DOI] [PubMed] [Google Scholar]
- 142.Danelich IM, Wright SS, Lose JM, Tefft BJ, Cicci JD, Reed BN. Safety of nonsteroidal antiinflammatory drugs in patients with cardiovascular disease. Pharmacother J Hum Pharmacol Drug Ther. 2015;35:520–35. doi: 10.1002/phar.1584. [DOI] [PubMed] [Google Scholar]
- 143.Konstantinopoulos PA, Lehmann DF. The cardiovascular toxicity of selective and nonselective cyclooxygenase inhibitors: comparisons, contrasts, and aspirin confounding. J Clin Pharmacol. 2005;45:742–50. doi: 10.1177/0091270005278202. [DOI] [PubMed] [Google Scholar]
- 144.Morita I. Distinct functions of COX-1 and COX-2. Prostaglandins Other Lipid Mediat. 2002;68:165–75. doi: 10.1016/s0090-6980(02)00029-1. [DOI] [PubMed] [Google Scholar]
- 145.Patrignani P, Sciulli MG, Manarini S, Santini G, Cerletti C, Evangelista V. COX-2 is not involved in thromboxane biosynthesis by activated human platelets. J Physiol Pharmacol. 1999;50:661–7. [PubMed] [Google Scholar]
- 146.Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M, DuBois RN. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell. 1998;93:705–16. doi: 10.1016/s0092-8674(00)81433-6. [DOI] [PubMed] [Google Scholar]
- 147.Shureiqi I, Chen D, Lee JJ, Yang P, Newman RA, Brenner DE, Lotan R, Fischer SM, Lippman SM. 15-LOX-1: a novel molecular target of nonsteroidal anti-inflammatory drug-induced apoptosis in colorectal cancer cells. J Natl Cancer Inst. 2000;92:1136–42. doi: 10.1093/jnci/92.14.1136. [DOI] [PubMed] [Google Scholar]
- 148.Ding XZ, Iversen P, Cluck MW, Knezetic JA, Adrian TE. Lipoxygenase inhibitors abolish proliferation of human pancreatic cancer cells. Biochem Biophys Res Commun. 1999;261:218–23. doi: 10.1006/bbrc.1999.1012. [DOI] [PubMed] [Google Scholar]
- 149.Ye YN, Wu WK, Shin VY, Bruce IC, Wong BC, Cho CH. Dual inhibition of 5-LOX and COX-2 suppresses colon cancer formation promoted by cigarette smoke. Carcinogenesis. 2005;26:827–34. doi: 10.1093/carcin/bgi012. [DOI] [PubMed] [Google Scholar]
- 150.Levy BD, De Sanctis GT, Devchand PR, Kim E, Ackerman K, Schmidt BA, Szczeklik W, Drazen JM, Serhan CN. Multi-pronged inhibition of airway hyper-responsiveness and inflammation by lipoxin A 4. Nat Med. 2002;8:1018–23. doi: 10.1038/nm748. [DOI] [PubMed] [Google Scholar]
- 151.Serhan CN, Levy B. Novel pathways and endogenous mediators in anti-inflammation and resolution. Chem Immunol Allergy. 2003;83:115–45. doi: 10.1159/000071558. [DOI] [PubMed] [Google Scholar]
- 152.Gaur U, Sahoo SK, De Tapas K, Ghosh PC, Maitra A, Ghosh PK. Biodistribution of fluoresceinated dextran using novel nanoparticles evading reticuloendothelial system. Int J Pharm. 2000;202:1–10. doi: 10.1016/s0378-5173(99)00447-0. [DOI] [PubMed] [Google Scholar]
- 153.Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev. 2012;64:206–12. doi: 10.1016/j.addr.2004.02.014. [DOI] [PubMed] [Google Scholar]
- 154.Amoozgar Z, Yeo Y. Recent advances in stealth coating of nanoparticle drug delivery systems. Wiley Interdiscip Rev Nanomedicine Nanobiotechnology. 2012;4:219–33. doi: 10.1002/wnan.1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Gregoriadis G, McCormack B. Targeting of drugs 6: strategies for stealth therapeutic systems. Vol. 300. Springer Science & Business Media; 2013. [Google Scholar]
- 156.Lasic DD, Martin FJ. Stealth liposomes. Vol. 20. CRC press; 1995. [Google Scholar]
- 157.Luxenhofer R, Sahay G, Schulz A, Alakhova D, Bronich TK, Jordan R, Kabanov AV. Structure-property relationship in cytotoxicity and cell uptake of poly (2-oxazoline) amphiphiles. J Control Release. 2011;153:73–82. doi: 10.1016/j.jconrel.2011.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Cheng G, Li G, Xue H, Chen S, Bryers JD, Jiang S. Zwitterionic carboxybetaine polymer surfaces and their resistance to long-term biofilm formation. Biomaterials. 2009;30:5234–40. doi: 10.1016/j.biomaterials.2009.05.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Terada T, Iwai M, Kawakami S, Yamashita F, Hashida M. Novel PEG-matrix metalloproteinase-2 cleavable peptide-lipid containing galactosylated liposomes for hepatocellular carcinoma-selective targeting. J Control Release. 2006;111:333–42. doi: 10.1016/j.jconrel.2005.12.023. [DOI] [PubMed] [Google Scholar]
- 160.Hu CM, Zhang L, Aryal S, Cheung C, Fang RH, Zhang L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci U S A. 2011;108:10980–5. doi: 10.1073/pnas.1106634108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Hatakeyama H, Akita H, Ito E, Hayashi Y, Oishi M, Nagasaki Y, Danev R, Nagayama K, Kaji N, Kikuchi H, Baba Y, Harashima H. Systemic delivery of siRNA to tumors using a lipid nanoparticle containing a tumor-specific cleavable PEG-lipid. Biomaterials. 2011;32:4306–16. doi: 10.1016/j.biomaterials.2011.02.045. [DOI] [PubMed] [Google Scholar]
- 162.Kopeček J, Kopečková P. HPMA copolymers: origins, early developments, present, and future. Adv Drug Deliv Rev. 2010;62:122–49. doi: 10.1016/j.addr.2009.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Metselaar JM, Bruin P, de Boer LW, de Vringer T, Snel C, Oussoren C, Wauben MH, Crommelin DJ, Storm G, Hennink WE. A novel family of L-amino acid-based biodegradable polymer− lipid conjugates for the development of long-circulating liposomes with effective drug-targeting capacity. Bioconjug Chem. 2003;14:1156–64. doi: 10.1021/bc0340363. [DOI] [PubMed] [Google Scholar]
- 164.Obata Y, Tajima S, Takeoka S. Evaluation of pH-responsive liposomes containing amino acid-based zwitterionic lipids for improving intracellular drug delivery in vitro and in vivo. J Control Release. 2010;142:267–76. doi: 10.1016/j.jconrel.2009.10.023. [DOI] [PubMed] [Google Scholar]
- 165.Park K, Lee SK, Park SA, Kim K, Chang HW, Jeong E, Park RW, Kim IS, Chan Kwon I, Byun Y, Kim SY. The attenuation of experimental lung metastasis by a bile acid acylated-heparin derivative. Biomaterials. 2007;28:2667–76. doi: 10.1016/j.biomaterials.2007.02.001. [DOI] [PubMed] [Google Scholar]
- 166.Rapoport NY, Kennedy AM, Shea JE, Scaife CL, Nam KH. Controlled and targeted tumor chemotherapy by ultrasound-activated nanoemulsions/microbubbles. J Control Release. 2009;138:268–76. doi: 10.1016/j.jconrel.2009.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ren TB, Xia WJ, Dong HQ, Li YY. Sheddable micelles based on disulfide-linked hybrid PEG-polypeptide copolymer for intracellular drug delivery. Polymer (Guildf) 2011;52:3580–6. [Google Scholar]
- 168.Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Deliv Rev. 2011;63:131–5. doi: 10.1016/j.addr.2010.03.011. [DOI] [PubMed] [Google Scholar]
- 169.Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv Drug Deliv Rev. 2013;65:71–9. doi: 10.1016/j.addr.2012.10.002. [DOI] [PubMed] [Google Scholar]
- 170.Maruyama K. Intracellular targeting delivery of liposomal drugs to solid tumors based on EPR effects. Adv Drug Deliv Rev. 2011;63:161–9. doi: 10.1016/j.addr.2010.09.003. [DOI] [PubMed] [Google Scholar]
- 171.Brown JM. Tumor hypoxia in cancer therapy. Methods Enzymol. 2007;435:295–321. doi: 10.1016/S0076-6879(07)35015-5. [DOI] [PubMed] [Google Scholar]
- 172.Brown JM, Wilson WR. Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 2004;4:437–47. doi: 10.1038/nrc1367. [DOI] [PubMed] [Google Scholar]
- 173.de Mendoza AE-H, Lasa-Saracibar B, Campanero MA, Blanco-Prieto M. Lipid nanoparticles in biomedicine. Encycl Nanosci Nanotechnol. 2010;15:455–78. [Google Scholar]
- 174.Hellsten R, Johansson M, Dahlman A, Sterner O, Bjartell A. Galiellalactone inhibits stem cell-like ALDH-positive prostate cancer cells. PLoS One. 2011;6:e22118. doi: 10.1371/journal.pone.0022118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Shi S, Yang K, Hong H, Chen F, Valdovinos HF, Goel S, Barnhart TE, Liu Z, Cai W. VEGFR targeting leads to significantly enhanced tumor uptake of nanographene oxide in vivo. Biomaterials. 2015;39:39–46. doi: 10.1016/j.biomaterials.2014.10.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Min KH, Kim JH, Bae SM, Shin H, Kim MS, Park S, Lee H, Park RW, Kim IS, Kim K, Kwon IC, Jeong SY, Lee DS. Tumoral acidic pH-responsive MPEG-poly (β-amino ester) polymeric micelles for cancer targeting therapy. J Control Release. 2010;144:259–66. doi: 10.1016/j.jconrel.2010.02.024. [DOI] [PubMed] [Google Scholar]
- 177.Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev. 2008;60:1615–26. doi: 10.1016/j.addr.2008.08.005. [DOI] [PubMed] [Google Scholar]
- 178.Sethuraman VA, Bae YH. TAT peptide-based micelle system for potential active targeting of anti-cancer agents to acidic solid tumors. J Control Release. 2007;118:21624. doi: 10.1016/j.jconrel.2006.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Zwicke GL, Ali Mansoori G, Jeffery CJ. Utilizing the folate receptor for active targeting of cancer nanotherapeutics. Nano Rev. 2012;3:18496. doi: 10.3402/nano.v3i0.18496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Choi CH, Alabi CA, Webster P, Davis ME. Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles. Proc Natl Acad Sci U S A. 2010;107:1235–40. doi: 10.1073/pnas.0914140107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Pawar SK, Badhwar AJ, Kharas F, Khandare JJ, Vavia PR. Design, synthesis and evaluation of N-acetyl glucosamine (NAG)-PEG-doxorubicin targeted conjugates for anticancer delivery. Int J Pharm. 2012;436:183–93. doi: 10.1016/j.ijpharm.2012.05.078. [DOI] [PubMed] [Google Scholar]
- 182.Patra M, Awuah SG, Lippard SJ. Chemical approach to positional isomers of glucose-platinum conjugates reveals specific cancer targeting through glucose-transporter-mediated uptake in vitro and in vivo. J Am Chem Soc. 2016;138:12541–51. doi: 10.1021/jacs.6b06937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Mitra D, Malkoski SP, Wang XJ. Cancer stem cells in head and neck cancer. Cancers (Basel) 2011;3:415–27. doi: 10.3390/cancers3010415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Zhao ZL, Zhang L, Huang CF, Ma SR, Bu LL, Liu JF, Yu GT, Liu B, Gutkind JS, Kulkarni AB, Zhang WF, Sun ZJ. NOTCH1 inhibition enhances the efficacy of conventional chemotherapeutic agents by targeting head neck cancer stem cell. Sci Rep. 2016;6:1–12. doi: 10.1038/srep24704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Merchant AA, Matsui W. Targeting Hedgehog-a cancer stem cell pathway. Clin cancer Res. 2010;16:3130–40. doi: 10.1158/1078-0432.CCR-09-2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–8. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
- 188.Chen K, Huang Y, Chen J. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin. 2013;34:732–40. doi: 10.1038/aps.2013.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Vinogradov S, Wei X. Cancer stem cells and drug resistance: the potential of nanomedicine. Nanomedicine. 2012;7:597–615. doi: 10.2217/nnm.12.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Landen CN, Goodman B, Katre AA, Steg AD, Nick AM, Stone RL, Miller LD, Mejia PV, Jennings NB, Gershenson DM, Bast RC Jr, Coleman RL, Lopez-Berestein G, Sood AK. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther. 2010;9:3186–99. doi: 10.1158/1535-7163.MCT-10-0563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Gao L, Yan L, Lin B, Gao J, Liang X, Wang Y, Liu J, Zhang S, Iwamori M. Enhancive effects of Lewis y antigen on CD44-mediated adhesion and spreading of human ovarian cancer cell line RMG-I. J Exp Clin Cancer Res. 2011;30:15. doi: 10.1186/1756-9966-30-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Borovski T, Felipe De Sousa EM, Vermeulen L, Medema JP. Cancer stem cell niche: the place to be. Cancer Res. 2011;71:634–9. doi: 10.1158/0008-5472.CAN-10-3220. [DOI] [PubMed] [Google Scholar]
- 193.Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, Kim JK, Patel LR, Ying C, Ziegler AM, Pienta MJ, Song J, Wang J, Loberg RD, Krebsbach PH, Pienta KJ, Taichman RS. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest. 2011;121:1298–312. doi: 10.1172/JCI43414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Hood JD, Bednarski M, Frausto R, Guccione S, Reisfeld RA, Xiang R, Cheresh DA. Tumor regression by targeted gene delivery to the neovasculature. Science. 2002;296:2404–7. doi: 10.1126/science.1070200. [DOI] [PubMed] [Google Scholar]
- 195.Fidler IJ. Angiogenesis and cancer metastasis. Cancer J. 2000;6:S134–41. [PubMed] [Google Scholar]
- 196.Scappaticci FA. Mechanisms and future directions for angiogenesis-based cancer therapies. J. Clin. Oncol. 2002;20:3906–27. doi: 10.1200/JCO.2002.01.033. [DOI] [PubMed] [Google Scholar]
- 197.Strieter RM, Belperio JA, Phillips RJ, Keane MP Seminars in Cancer Biology. CXC chemokines in angiogenesis of cancer. Elsevier; 2004. pp. 195–200. [DOI] [PubMed] [Google Scholar]
- 198.Reynolds AR, Moghimi SM, Hodivala-Dilke K. Nanoparticle-mediated gene delivery to tumour neovasculature. Trends Mol Med. 2003;9:2–4. doi: 10.1016/s1471-4914(02)00004-7. [DOI] [PubMed] [Google Scholar]
- 199.Ribatti D, Annese T, Ruggieri S, Tamma R, Crivellato E. Limitations of anti-angiogenic treatment of tumors. Transl Oncol. 2019;12:981–6. doi: 10.1016/j.tranon.2019.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, Hahn SM, Hamblin MR, Juzeniene A, Kessel D, Korbelik M, Moan J, Mroz P, Nowis D, Piette J, Wilson BC, Golab J. Photodynamic therapy of cancer: an update. CA Cancer J Clin. 2011;61:250–81. doi: 10.3322/caac.20114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Li X, Kolemen S, Yoon J, Akkaya EU. Activatable photosensitizers: agents for selective photodynamic therapy. Adv Funct Mater. 2017;27:1604053. [Google Scholar]
- 202.Dolmans DE, Fukumura D, Jain RK. Photodynamic therapy for cancer. Nat Rev Cancer. 2003;3:380–7. doi: 10.1038/nrc1071. [DOI] [PubMed] [Google Scholar]
- 203.Majumdar P, Nomula R, Zhao J. Activatable triplet photosensitizers: magic bullets for targeted photodynamic therapy. J Mater Chem C. 2014;2:5982–97. [Google Scholar]
- 204.Abrahamse H, Hamblin MR. New photosensitizers for photodynamic therapy. Biochem J. 2016;473:347–64. doi: 10.1042/BJ20150942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Hamblin MR, Hasan T. Photodynamic therapy: a new antimicrobial approach to infectious disease? Photochem Photobiol Sci. 2004;3:436–50. doi: 10.1039/b311900a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour immunity. Nat Rev Cancer. 2006;6:535–45. doi: 10.1038/nrc1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Lovell JF, Liu TW, Chen J, Zheng G. Activatable photosensitizers for imaging and therapy. Chem Rev. 2010;110:2839–57. doi: 10.1021/cr900236h. [DOI] [PubMed] [Google Scholar]
- 208.Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther. 2006;5:1275–9. doi: 10.1158/1535-7163.MCT-06-0024. [DOI] [PubMed] [Google Scholar]
- 209.Lau JT, Lo PC, Jiang XJ, Wang Q, Ng DK. A dual activatable photosensitizer toward targeted photodynamic therapy. J Med Chem. 2014;57:4088–97. doi: 10.1021/jm500456e. [DOI] [PubMed] [Google Scholar]
- 210.Cheng R, Feng F, Meng F, Deng C, Feijen J, Zhong Z. Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery. J Control Release. 2011;152:2–12. doi: 10.1016/j.jconrel.2011.01.030. [DOI] [PubMed] [Google Scholar]
- 211.Kim H, Kim Y, Kim IH, Kim K, Choi Y. ROS-responsive activatable photosensitizing agent for imaging and photodynamic therapy of activated macrophages. Theranostics. 2014;4:1. doi: 10.7150/thno.7101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Lee MH, Kim JY, Han JH, Bhuniya S, Sessler JL, Kang C, Kim JS. Direct fluorescence monitoring of the delivery and cellular uptake of a cancer-targeted RGD peptide-appended naphthalimide theragnostic prodrug. J Am Chem Soc. 2012;134:12668–74. doi: 10.1021/ja303998y. [DOI] [PubMed] [Google Scholar]
- 213.Lagendijk JJ. Hyperthermia treatment planning. Phys Med Biol. 2000;45:R61. doi: 10.1088/0031-9155/45/5/201. [DOI] [PubMed] [Google Scholar]
- 214.Kok HP, Wust P, Stauffer PR, Bardati F, Van Rhoon GC, Crezee J. Current state of the art of regional hyperthermia treatment planning: a review. Radiat Oncol. 2015;10:196. doi: 10.1186/s13014-015-0503-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Kasevich RS, McQueeney JF, Crooker RH, inventors. Method and apparatus for hyperthermia treatment. Google Patents. 1988
- 216.Kobayashi T, Ito A, Honda H Hyperthermic Oncology from Bench to Bedside. Magnetic nanoparticle-mediated hyperthermia and induction of anti-tumor immune responses. Springer; 2016. pp. 137–50. [Google Scholar]
- 217.Schirrmacher V, Lorenzen D, Van Gool SW, Stuecker W. A new strategy of cancer immunotherapy combining hyperthermia/oncolytic virus pretreatment with specific autologous anti-tumor vaccination-A review. Austin Oncol Case Rep. 2017;2:1006. [Google Scholar]
- 218.Weigelin B, Krause M, Friedl P. Cytotoxic T lymphocyte migration and effector function in the tumor microenvironment. Immunol Lett. 2011;138:19–21. doi: 10.1016/j.imlet.2011.02.016. [DOI] [PubMed] [Google Scholar]
- 219.Colombo R, Salonia A, Leib Z, Pavone-Macaluso M, Engelstein D. Long-term outcomes of a randomized controlled trial comparing thermochemotherapy with mitomycin-C alone as adjuvant treatment for non-muscle-invasive bladder cancer (NMIBC) BJU Int. 2011;107:912–8. doi: 10.1111/j.1464-410X.2010.09654.x. [DOI] [PubMed] [Google Scholar]
- 220.van Haaren PM, Kok HP, van den Berg CA, Zum Vörde Sive Vörding PJ, Oldenborg S, Stalpers LJ, Schilthuis MS, de Leeuw AA, Crezee J. On verification of hyperthermia treatment planning for cervical carcinoma patients. Int J Hyperth. 2007;23:303–14. doi: 10.1080/02656730701297538. [DOI] [PubMed] [Google Scholar]
- 221.Franckena M, Fatehi D, de Bruijne M, Canters RA, van Norden Y, Mens JW, van Rhoon GC, van der Zee J. Hyperthermia dose-effect relationship in 420 patients with cervical cancer treated with combined radiotherapy and hyperthermia. Eur J Cancer. 2009;45:1969–78. doi: 10.1016/j.ejca.2009.03.009. [DOI] [PubMed] [Google Scholar]
- 222.Sreenivasa G, Gellermann J, Rau B, Nadobny J, Schlag P, Deuflhard P, Felix R, Wust P. Clinical use of the hyperthermia treatment planning system HyperPlan to predict effectiveness and toxicity. Int J Radiat Oncol Biol Phys. 2003;55:407–19. doi: 10.1016/s0360-3016(02)04144-5. [DOI] [PubMed] [Google Scholar]
- 223.Hand JW, Machin D, Vernon CC, Whaley JB. Analysis of thermal parameters obtained during phase III trials of hyperthermia as an adjunct to radiotherapy in the treatment of breast carcinoma. Int J Hyperth. 1997;13:343–64. doi: 10.3109/02656739709046538. [DOI] [PubMed] [Google Scholar]
- 224.Elliott JC, inventor. Hyperoxygenation/hyperthermia treatment apparatus. Google Patents. 2019
- 225.Hand JW, Lau RW, Lagendijk JJ, Ling J, Burl M, Young IR. Electromagnetic and thermal modeling of SAR and temperature fields in tissue due to an RF decoupling coil. Magn Reson Med. 1999;42:183–92. doi: 10.1002/(sici)1522-2594(199907)42:1<183::aid-mrm24>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- 226.Deuflhard P, Schiela A, Weiser M. Mathematical cancer therapy planning in deep regional hyperthermia. Acta Numer. 2012;21:307–78. [Google Scholar]
- 227.Lollini PL, Cavallo F, Nanni P, Forni G. Vaccines for tumour prevention. Nat Rev Cancer. 2006;6:204–16. doi: 10.1038/nrc1815. [DOI] [PubMed] [Google Scholar]
- 228.Mitchell MS. Cancer vaccines, a critical review--Part I. Curr Opin Investig Drug. 2002;3:140–9. [PubMed] [Google Scholar]
- 229.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12:265–77. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Mitchell MS. Cancer vaccines, a critical review--Part II. Curr Opin Investig Drugs. 2002;3:150–8. [PubMed] [Google Scholar]
- 231.van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer. 2016;16:219. doi: 10.1038/nrc.2016.16. [DOI] [PubMed] [Google Scholar]
- 232.Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005;5:296–306. doi: 10.1038/nri1592. [DOI] [PubMed] [Google Scholar]
- 233.Babu RSA, Kumar KK, Reddy GS, Anuradha C. Cancer vaccine: a review. J Orofac Sci. 2010;2:77. [Google Scholar]
- 234.Frazer IH. Prevention of cervical cancer through papillomavirus vaccination. Nat Rev Immunol. 2004;4:46–55. doi: 10.1038/nri1260. [DOI] [PubMed] [Google Scholar]
- 235.Allen UD. Immunizations for children with cancer. Pediatr Blood Cancer. 2007;49:1102–8. doi: 10.1002/pbc.21346. [DOI] [PubMed] [Google Scholar]
- 236.McNamara MA, Nair SK, Holl EK. RNA-based vaccines in cancer immunotherapy. J Immunol Res. 2015;2015:794528. doi: 10.1155/2015/794528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Vormehr M, Türeci Ö, Sahin U. Harnessing tumor mutations for truly individualized cancer vaccines. Annu Rev Med. 2019;70:395–407. doi: 10.1146/annurev-med-042617-101816. [DOI] [PubMed] [Google Scholar]
- 238.Türeci Ö, Löwer M, Schrörs B, Lang M, Tadmor A, Sahin U. Challenges towards the realization of individualized cancer vaccines. Nat Biomed Eng. 2018;2:566–9. doi: 10.1038/s41551-018-0266-2. [DOI] [PubMed] [Google Scholar]
- 239.Nanni P, Nicoletti G, Palladini A, Croci S, Murgo A, Antognoli A, Landuzzi L, Fabbi M, Ferrini S, Musiani P, Iezzi M, De Giovanni C, Lollini PL. Antimetastatic activity of a preventive cancer vaccine. Cancer Res. 2007;67:11037–44. doi: 10.1158/0008-5472.CAN-07-2499. [DOI] [PubMed] [Google Scholar]