repurposing existing drugs with anti-cancer effects

Drug Repurposing in Cancer

Drug Repurposing in Cancer

Last Section Update: 08/2022

Contributor(s): Maureen Williams, ND; Shayna Sandhaus, PhD

1 Introduction

Efforts to develop new cancer therapies are costly and not always successful. Therefore, repurposing existing drugs with potential anti-cancer effects to expand the armamentarium of cancer therapeutics is an appealing strategy. Common drugs that show promise as repurposed adjuvant cancer therapies are often inexpensive and have well-documented safety and drug-interaction profiles.1

Given the appeal of established and well-characterized drugs from a cost and safety perspective, several research collaborations have emerged to study the potential of repurposed drugs for use in cancer. Among the most well-known of these collaborative efforts is the Repurposing Drugs in Oncology (ReDO) Project, which was launched by an international group of researchers in 2014.2 More recently, high-throughput computer-aided drug screening efforts continue to reveal the potential of repurposing existing drugs as cancer therapeutics.3

A major challenge in advancing research on repurposed drugs for cancer is that there is often little commercial interest in this strategy from pharmaceutical companies. Repurposing old drugs is not as financially lucrative as developing new drugs and bringing them to market. Therefore, research on repurposing existing drugs gets little financial support from the private sector. Some support for drug repurposing research is provided through federal funding in the United States: both the National Institutes of Health (NIH) and the Food and Drug Administration (FDA) have drug repurposing programs.4

The purpose of this protocol is to highlight several drugs with track records of safe use in other conditions that may have utility in the context of cancer. The drugs discussed in this protocol should not be incorporated into any cancer treatment regimen unless done so under the supervision of qualified oncology-care clinicians.

Note: Refer also to Life Extension’s other cancer-related protocols, including Cancer Adjuvant Therapy, Chemotherapy, Cancer Radiation Therapy, Cancer Surgery, Cancer Treatment: The Critical Factors, and the protocol on the specific type of cancer you are interested in.

Understanding the Evidence Presented in this Protocol

The evidence presented in this protocol is generally categorized as either correlational or causal evidence.

Correlational evidence comes from observational studies that cannot establish a cause-and-effect relationship between the drug in question and the outcomes being studied. Conversely, in medicine, causal evidence comes from randomized controlled clinical trials. Causal evidence from clinical trials is considered the “gold standard” in medicine because it is generally less confounded and biased than observational evidence. However, not all clinical trials are equal—small trials with weak designs generate weak evidence.

Unfortunately, many of the clinical trials conducted to investigate drug repurposing are low quality, smaller studies. This means the overall strength of the evidence is often not robust for several of the drugs discussed in this protocol. This limitation of evidence quality should be kept in mind while reading this protocol.

2 Metformin

Background

Metformin (Glucophage) is a medication used to treat diabetes, and has been shown to reduce the progression from pre-diabetes to overt diabetes.5 After metformin was first approved by the FDA in late 1994, evidence emerged suggesting people taking metformin for diabetes were less likely to develop certain types of cancer.6,7 Subsequent studies showed that metformin use was associated with improved survival in diabetic patients with several different types of cancer.6 These unexpected observations may be explained by cellular effects of metformin that have since come to light.

Metformin decreases blood glucose levels by inhibiting glucose production and release by the liver; in addition, metformin increases cells’ sensitivity to insulin. These effects can reduce high insulin levels in people with insulin resistance and type 2 diabetes.8 Because cancer cells have high glucose requirements and insulin can promote tumor initiation and progression, lower glucose and insulin levels may protect against cancer development and growth.9,10

Metformin has also been shown to affect signaling pathways that influence the activity, growth, and survival of cancer cells. Although its exact mechanisms are not completely understood, metformin’s actions are likely due in part to suppression of mitochondrial energy production and activation of AMP-activated protein kinase (AMPK), an enzyme that slows down cellular energy use and regulates cell growth and proliferation.7,11-13 In addition, metformin has been shown to inhibit cell signaling via a pathway known as mammalian target of rapamycin (mTOR). Signaling via the mTOR pathway is involved in regulating cell division and survival, and dysregulated mTOR signaling is a common characteristic of cancer cells. Studies in cancer cells suggest metformin-induced AMPK activation and mTOR inhibition can inhibit cancer cell initiation, proliferation, and metastasis, and induce apoptosis (normal cell death).6,7,9,12 Metformin also impacts the tumor microenvironment, reducing inflammatory immune function, quieting tumor-supportive regulatory T cells, and stimulating anti-cancer T cells.11,14,15

In addition, metformin has been shown to modulate the microbiome in ways that may mitigate inflammation and confer some protection against cancer. In the context of colorectal cancer, for example, researchers have recently proposed a “metformin-gut microbiota-colorectal cancer (in type 2 diabetes) axis.”16 In this new theoretical framework, “the gut microbiota plays a key role as a bridge that affects not only the pharmacological effects of metformin but also the occurrence of colorectal cancer” in patients with type 2 diabetes. This model proposes that metformin’s inhibition of colorectal cancer in people with type 2 diabetes may be mainly due to manipulation of populations of several microbiota genera in the gut, including Bacteroides, Ruminococcus, Clostridium, Firmicute, Lactobacillus, and E. coli. More research is needed, but evidence available as of early 2021 increasingly suggests manipulation of the gut microbiome may be a principal mode of action for metformin’s influence on metabolic activity and an important component of the drugs’ anti-cancer actions.17-20

Observational (Correlational) Evidence

The growing body of research is varied in methodologies and definitions, such as how metformin use is defined and what characterizes the control group; therefore, more research is needed to clarify metformin’s potential as an adjuvant therapy for cancer. The following summary describes recent findings from observational studies of the effects of metformin in the context of specific types of cancer in people with type 2 diabetes.

Metformin and colorectal cancer: A meta-analysis of 50 studies with a combined total of more than 238,000 participants found metformin use was associated with lower risk of colorectal cancer and longer overall and cancer-specific survival in patients with type 2 diabetes. Greater protective effects were seen with higher doses and longer duration of use in Asian populations.21 Findings from another meta-analysis suggest women may benefit more than men.22 In other studies in diabetic colorectal cancer patients, metformin use was linked to better responsiveness to chemotherapy and radiation therapy, a lower rate of cancer recurrence, fewer metastases, and longer survival.23,24

Metformin and breast cancer: Although there have been conflicting findings, some observational studies have reported lower breast cancer incidence and mortality in type 2 diabetic patients treated with metformin than in those not treated with metformin, and the benefits may pertain particularly to hormone receptor-positive and HER2-positive breast cancers.658-660 However, randomized controlled trials and meta-analyses have shown metformin is not likely to improve outcomes in non-diabetic breast cancer patients.661-663

Metformin and ovarian cancer: A meta-analysis of five studies with a combined total of 3,582 ovarian cancer patients found consistent evidence for improved survival in those using metformin as an anti-diabetes treatment after their cancer diagnosis compared with both diabetic and non-diabetic non-metformin users. Two other meta-analyses that each included 13 studies found an association between metformin use and lower ovarian cancer incidence, as well as a link between metformin use and improved prognosis in ovarian cancer patients.26,27 However, one meta-analysis of six studies with 2,638 participants reached another conclusion, finding no correlation between metformin use and ovarian cancer outcomes.28

Metformin and liver cancer: A meta-analysis of eight studies found metformin use was correlated with reduced risk of hepatocellular carcinoma (the most common type of primary liver cancer) in type 2 diabetic subjects.29 In another meta-analysis that included results from six studies in diabetic patients with hepatocellular carcinoma, the pooled data indicated metformin use was related to increased overall and recurrence-free survival in those treated with measures intended to cure the cancer; however, metformin had no association with survival in those whose cancer treatment was palliative.30 A retrospective cohort study of 164 individuals with diabetes and single nodular hepatocellular carcinoma who underwent transarterial chemoembolization (TACE) found those who took metformin (median duration 32 months and median dosage 250 mg daily) had significantly higher response rates—79% versus 60% in those who did not take metformin. Metformin use was found to be an independent predictor of response rates to TACE and was associated with a 72% lower risk of local tumor recurrence. Although metformin use prior to TACE reduced local tumor recurrence, its use did not appear to affect overall recurrence rate or progression-free survival.684

Metformin and pancreatic cancer: Metformin use has been linked to reduced risk of pancreatic cancer.31 In a case-control study based at the University of Texas MD Anderson Cancer Center in Houston, diabetic patients with a history of metformin use had considerably lower odds of pancreatic cancer than those who had not taken metformin (OR: 0.38, 95% CI: 0.22-0.69). On the other hand, diabetic patients in this study who had taken insulin or insulin secretagogues had significantly greater odds of pancreatic cancer.32 A meta-analysis of 14 studies including nearly 36,800 participants with type 2 diabetes and pancreatic cancer found metformin use was correlated with increased overall survival, with stronger effects in those with less-advanced tumors. A second analysis of the data, however, showed the protective effect was only present in Asians.33 Another study found no benefit from metformin in diabetic African Americans with pancreatic cancer; however, in diabetic Caucasian patients who started metformin after the time of cancer diagnosis, an increase in overall survival was observed.34

Metformin and gastric cancer: A meta-analysis of findings from 11 studies found metformin use was associated with a reduced risk of gastric cancer and longer cancer-specific and overall survival in type 2 diabetics in Asian, but not Western, populations.35 A study done in the Baltic country of Lithuania found, compared with the general population, that people using metformin for type 2 diabetes had lower incidence of gastric cancer, while those using sulfonylureas (another type of anti-diabetes drug) had a higher incidence.36

Metformin and lung cancer: Lithuanian researchers reported the risk of death was reduced in lung cancer patients with diabetes being treated with metformin, but was increased in those treated with sulfonylureas, compared with non-diabetic lung cancer patients.37 A small study performed in the United States found, in 50 patients with non-small cell lung cancer, those simultaneously receiving metformin for diabetes had better cancer treatment response, better disease control, and longer progression-free and overall survival compared with non-diabetics.38 An observational registry study that assessed data from over 730,000 subjects found that metformin use was associated with lower lung cancer incidence and mortality. Moreover, increasing metformin dose was associated with more pronounced reductions in lung cancer risk in this study. This study also showed that combined use of metformin, statins, and aspirin was associated with greater reductions in lung cancer mortality than use of any of the three alone; this effect was greater with longer duration use.39

Metformin and prostate cancer: Several meta-analyses have found metformin use does not significantly influence prostate cancer risk40-42; however, in one meta-analysis, diabetic prostate cancer patients using metformin were found to have prolonged cancer-specific, recurrence-free, and overall survival.41

Metformin and brain cancer: In patients with high-grade glioma (the most common type of brain cancer), metformin use was associated with longer progression-free and overall survival in those with grade III but not grade IV disease.43 (Grade IV is the most malignant form of glioma; it is also known as glioblastoma.) In addition, a meta-analysis of three studies involving newly diagnosed glioblastoma patients found no benefit associated with metformin use.44

Metformin and endometrial cancer: Findings from meta-analyses suggest there is no relationship between metformin use and endometrial cancer risk,45-47 but metformin use has been consistently found to be associated with better survival in endometrial cancer patients.47-51

Metformin and blood cancers: In Taiwanese people with type 2 diabetes, metformin use was associated with lower risk of non-Hodgkin lymphoma compared with other diabetes treatments52; however, a similar study done in Canada found no relationship.53 In addition, metformin use does not appear to correlate with improved outcomes in patients with non-Hodgkin lymphoma.54,55 In people with a precancerous condition called monoclonal gammopathy of undetermined significance (MGUS), those treated with metformin were found to have slower progression from MGUS to multiple myeloma.56,57 In addition, some evidence suggests metformin use may be linked to better outcomes in diabetics with multiple myeloma.58

Metformin and other cancers: A study done in Sweden found metformin users had a lower risk of esophageal cancer than non-metformin users.59 In a Taiwanese study, people with type 2 diabetes were less likely to be diagnosed with esophageal cancer than those treated with other medications.60 A meta-analysis of seven studies found metformin use was unrelated to survival in patients with head and neck cancer.61

Causal Evidence - Clinical Trial Data

Recent randomized clinical trials have begun to examine whether metformin can also help non-diabetic cancer patients:

  • Among 200 women with an early form of breast cancer, a marker of tumor aggressiveness and potential for proliferation was significantly reduced in breast tissue surrounding tumors after 28 days of treatment with 1,700 mg per day of metformin compared with placebo.62
  • In 58 women with HER2-positive breast cancer treated with standard chemotherapy, the addition of 1,700 mg of metformin for 24 weeks increased the likelihood of a complete response63; in addition, even those who did not have a complete response expressed lower amounts of a marker of tumor aggressiveness.64
  • In 44 men with treatment-resistant prostate cancer, treatment with 1,000 mg per day of metformin appeared to stabilize their cancer and slow the increase of prostate-specific antigen (PSA) levels, a tumor marker used to monitor prostate cancer growth.65
  • In 18 patients with advanced or metastatic non-small cell lung cancer who received metformin at doses of 1,000 to 2,000 mg per day along with standard chemotherapy for one year, 47% experienced no cancer progression. This rate of non-progression was better than the historical rate of about 15% seen in people with this type of cancer.66
  • A controlled trial in 139 patients with advanced lung cancer found adding 500 mg of metformin twice daily to chemotherapy resulted in longer progression-free survival.67
  • In 102 patients with acute lymphoblastic leukemia (ALL), 850 mg of metformin three times daily improved the response to chemotherapy and reduced the rate of relapse compared with chemotherapy alone. Furthermore, this trial identified a genetic marker associated with poorer outcomes in general but greater benefit from metformin.68
  • In a placebo-controlled trial, 151 individuals with recurrent precancerous colon polyps were treated surgically and then given low-dose metformin (250 mg per day) or placebo for one year; those who received metformin had fewer benign and precancerous polyps at 1-year follow-up colonoscopy.69
  • A small early clinical trial in ovarian cancer patients reported no improvement in progression-free survival with the addition of metformin to chemotherapy.70

More studies are underway to determine optimal doses of metformin, define which types of cancer and types of patients are most responsive to metformin, and test the effects of metformin in combinations with radiation therapy and chemotherapy.71

Safety Considerations

Metformin may cause side effects such as gastrointestinal upset including bloating, flatulence, and diarrhea.72 Uncommonly, it may also cause lactic acidosis (high levels of lactic acid), particularly when used in high doses.71,73 Metformin may raise homocysteine concentrations as well, but supplementation with B vitamins may counteract this side effect.74 Metformin may reduce testosterone levels as well (which may be beneficial among women with polycystic ovary syndrome).75-77 In addition, patients with impaired kidney function and elderly individuals with heart failure or other serious conditions should be closely monitored while using metformin.9,78

3 Statin Drugs

Background

Statins are a family of cholesterol-lowering drugs that work by inhibiting an enzyme called HMG-CoA reductase, which is critical to cholesterol synthesis in the liver. There are currently seven drugs in the statin family approved for treating high cholesterol levels: simvastatin (Zocor), lovastatin (Mevacor), pravastatin (Pravachol), fluvastatin (Lescol), atorvastatin (Lipitor), rosuvastatin (Crestor), and pitavastatin (Livalo). They differ somewhat in degree of lipid-solubility, metabolism, and clearance, and have different effects on lipid profiles.79

Cholesterol is needed for cell membrane structure, energy storage, and as a substrate for signaling molecules. Cancer cells, especially cancer stem cells, are dependent on the cholesterol-synthesis pathway to support proliferation, migration, and invasion. By interfering with cholesterol synthesis, statins may therefore suppress cancer activity.80-82 There is also evidence that statins lower immune signaling and help regulate epigenetic mechanisms related to cells’ proliferative potential. Because their effects on HMG-CoA reductase impact multiple fundamental cellular processes, statins are considered pleiotropic agents.83,84

In a meta-analysis of seven studies with a combined total of 23,555 participants, all of whom had type 2 diabetes, cancer incidence was found to be lower in those taking any of the statin medications, with rosuvastatin showing the strongest association.85 Another meta-analysis reported people taking statins for high cholesterol levels had lower incidences of 10 out of 18 types of cancer, although the evidence for these correlations was mostly weak.86 Although not all studies agree,87 findings from dozens of studies and numerous research reviews suggest the incidental use of statins to lower high cholesterol levels may reduce cancer risk and, in cancer patients, prolong survival and lower rates of cancer recurrence.88-90

Despite many positive findings, much remains to be learned about the relationship between statin use and cancer initiation and progression. For example, there is conflicting evidence for cancer-related benefits with long-term statin use: while multiple studies suggest a protective effect, a recent meta-analysis of 15 studies with a total of 358,544 participants found those taking statins for a minimum of 10 years experienced no benefit on cancer incidence or cancer-related mortality.91 Importantly, some evidence suggests long-term statin use may be associated with new-onset diabetes.92 More research is needed to clarify the risk-benefit profile in terms of potential cancer risk reduction or treatment benefit against the potential detrimental effect of long-term statin use on glucose and insulin metabolism. Statins have a well-documented favorable risk-benefit in the context of cardiovascular risk, but more evidence is needed to establish their utility in the prevention and/or treatment of cancer in otherwise healthy individuals, especially with long-term use.

Observational (Correlational) Evidence

Statins and liver cancer: Statin users have consistently been found to have a substantially lower incidence of hepatocellular carcinoma, the most common form of cancer originating in the liver.93-95 A meta-analysis of data from 24 studies with a total of 59,073 participants found statin use was associated with a 46% hepatocellular carcinoma risk reduction, and the association was even stronger in patients with increased liver cancer risk due to diabetes, cirrhosis, or viral hepatitis.96 In one large study, the correlation between statin use and reduced liver cancer risk was only evident in those taking highly lipid-soluble statins (atorvastatin, fluvastatin, lovastatin, pitavastatin, and simvastatin) and the reduction was greatest in those who took a cumulative total of 600 or more daily doses. In addition, 10-year mortality was reduced in users of all types of statins compared with non-users.97

Statins and esophageal cancer: A large meta-analysis examined data from 22 studies that followed a total of more than 6 million people and identified 372,206 esophageal cancer cases. The analysis found statin users, including those at high risk due to Barrett’s esophagus (a precancerous condition of the esophagus), had a lower incidence of esophageal cancer.98 A meta-analysis of data from four studies with a total of 20,435 esophageal cancer patients found statin use after cancer diagnosis was correlated with better cancer-specific and overall survival.99

Statins and gastric cancer: A relationship between long-term statin use and reduced gastric cancer risk was reported in a Korean study that included 21,149 participants with high cholesterol levels and followed them for 12–13 years.100 In another Korean study that included people at high risk of gastric cancer due to Helicobacter pylori, those taking statins were less likely to develop gastric cancer after H. pylori treatment compared with those not taking statins.101 Another Korean study found an association between statin use and reduced recurrence rate during 5–12 years of follow-up in gastric cancer patients treated surgically.102 An analysis of two populations in the United Kingdom, however, found evidence for a modest benefit of statins on risk of gastric cancer.103

Statins and pancreatic cancer: Despite some conflicting evidence,104 statin use was found by two large meta-analyses to be correlated with reduced risk of pancreatic cancer.105,106 Statin use may also be associated with lower incidence of pancreatic cancer in patients at high risk due to chronic pancreatitis,107,108 though not all studies agree.109 There is also evidence suggesting statins may improve the course of pancreatic cancer. A meta-analysis of 14 studies with a combined total of 33,137 pancreatic cancer patients found statin use was associated with a lower mortality rate in those treated surgically, but not those with advanced cancer. Some data in the analysis suggested rosuvastatin specifically was linked to better survival.110 In an analysis of two studies comparing outcomes in a combined total of 156 pancreatic cancer patients who used statins during cancer treatment with 641 patients who did not, statin use was associated with better progression-free and overall survival.111 Regular statin use prior to cancer diagnosis was also associated with a small increase in survival time in pancreatic cancer patients participating in the Nurses’ Health Study and the Health Professionals’ Follow-Up Study.112

Statins and prostate cancer: Long-term statin use (10 years or longer) has also been associated with reduced prostate cancer risk in some, but not all, studies.113,114 While some research suggests statins may protect against aggressive prostate tumors, metastatic disease, and mortality,115,116 studies in men with elevated PSA levels or those under active surveillance for low-risk tumors have not demonstrated a link between statin use and prostate cancer occurrence or progression.117,118 Statin use following prostate cancer diagnosis has been correlated with better survival outcomes in multiple observational studies.119-122 For example, a study that followed 44,126 men for 24 years found no relationship between statin use and prostate cancer risk, but among men who developed prostate cancer, those using statins at the time of cancer diagnosis were less likely to die from their cancer than those who never used statins or had stopped using them prior to their cancer diagnosis. The study also identified an epigenetically controlled marker on tumor cells that may indicate responsiveness to statin therapy.123 One study found relationships between statin use and improved survival outcomes in those taking statins for 11 months or longer or who had taken a higher cumulative dose of statins, and in those with more aggressive tumors.124

Statins and breast cancer: Although statins do not appear to impact the overall risk of breast cancer, there is some evidence that statin use may reduce breast cancer-related mortality.125,126 Findings from a meta-analysis of data from seven studies, including a combined total of 197,048 breast cancer patients, suggest statins with high lipid solubility (atorvastatin, fluvastatin, lovastatin, pitavastatin, and simvastatin) are more strongly associated with increased survival than those with low lipid solubility (rosuvastatin and pravastatin).127 In one study, disease-free and overall survival were higher in breast cancer patients who had taken statins for five years or longer.128 A study performed in Sweden evaluated data on statin use among 20,559 breast cancer patients. Women who took statins regularly within the six months prior to their cancer diagnosis had a lower risk of breast cancer-related death during approximately five years of monitoring compared with those who never or irregularly used statins during the six months prior to diagnosis. In addition, those who initiated statin use after their cancer diagnosis also had a lower risk of breast cancer-related death.129 In breast cancer survivors, statin use was associated with reduced risk of new cancer in the other breast, and the association was stronger with long-term (five years or longer) use.130 However, according to a retrospective cohort study, statin use may not influence breast cancer risk in women with type 2 diabetes.131

Statins and lung cancer: Long-term statin use has been observed to be correlated with lower lung cancer risk in Korean men, particularly smokers, as well as in a Taiwanese population with chronic obstructive pulmonary disease (COPD).132,133 In addition, two large meta-analyses found statin use before or during cancer treatment was associated with lower cancer-specific and all-cause mortality, particularly in patients with non-small cell lung cancer and those with more advanced cancer.134,135 However, statin use did not appear to impact survival in patients who underwent surgery for lung cancer.136

Statins and colorectal cancer: Studies of the effect of statin drugs on colorectal cancer risk and outcomes have produced varying results ranging from some benefit to no effect.140,143,650-654 Variables that may influence results include differences in dosage; pharmacological differences among different statin drugs; treatment history (eg, duration and initiation before vs. after diagnosis); tumor-specific factors like location and genetics; and comorbid conditions such as diabetes, inflammatory bowel disease, and cardiovascular disease.140,145,652,655

One example of statins’ potential benefit in colorectal cancer patients comes from a retrospective cohort study that included nearly 12,000 patients who underwent surgical resection for rectal cancer. This study found that statin use within the year before surgery was significantly associated with a reduced risk of 90-day all-cause mortality (0.7% with statins vs. 5.5% without statins).656

As of 2021 the American Gastroenterological Association recommends statins not be used to prevent colorectal cancer in those at average risk, and that statins should not be prescribed to reduce mortality in colorectal cancer patients.657

Statins and endometrial cancer: While statin use does not appear to be correlated with endometrial cancer risk in the general population,146,147 it may have a protective effect against endometrial cancer in women with type 2 diabetes.148,149 In addition, a meta-analysis of data from nine studies found statins had a positive impact on overall and disease-specific survival.32 In one study with 6,694 participants, those who used statins before and after endometrial cancer diagnosis had a higher chance of survival, with a more pronounced association more than five years after diagnosis compared with years 1 through 5 after diagnosis.150 Not all studies agree, however: a study that included data from 2,017 endometrial cancer patients found no relationship between statin use and recurrence-free or overall survival during an average follow-up time of 6.2 years.151

Statins and ovarian cancer: Although not all studies have linked statin use to lower ovarian cancer risk,146,152 one study that followed 2,040 women with and 2,100 without ovarian cancer for 16 years found statin use for six months or longer was more common in women who did not develop ovarian cancer than those who did.153 There is also evidence statins may improve the course of ovarian cancer. In a study that included data from 2,195 women with ovarian cancer monitored for an average of 2.2 years, cancer-related mortality was lower in those who began taking a statin within one year after their cancer diagnosis.154 A meta-analysis of eight other studies with a combined total of 19,904 ovarian cancer patients also noted improved survival in those who used statins after their cancer diagnosis.155

Statins and other cancers: There is also evidence that statin use is associated with lower incidence and better survival rates in head and neck cancer,156,157 lower risk and better survival rates in kidney cancer,158-160 and lower incidences of blood cancers including chronic lymphocytic leukemia, non-Hodgkin lymphoma, and multiple myeloma.161-163 However, statin use does not appear to be correlated with lower risk of, or better treatment outcomes for, brain cancer.164-166

Causal Evidence - Clinical Trial Data

Despite intriguing preclinical research suggesting statins have pleotropic effects that may be beneficial in the context of cancer,167 prospective clinical trials have not suggested statin use reduces cancer risk or provides benefit as adjunctive cancer therapy. A meta-analysis of 10 randomized controlled trials in patients with advanced cancers found adding statins to cancer treatment had no impact on survival outcomes.168 In a meta-analysis of eight randomized, controlled, phase II and III trials, adding statins at typical doses to cancer treatment was not found to improve outcomes in patients with solid tumors including liver, gastric, lung, prostate, and colorectal cancers.169 Another meta-analysis of 21 randomized controlled trials involving over 65,000 subjects found that “… there was no significant association between statin use and risk of cancer (RR: 0.97; 95% CI: 0.92-1.02).170 Most trials included in these meta-analyses used standard cholesterol-lowering doses of either simvastatin or pravastatin; at least one of the included trials used lovastatin. Whether use of higher doses or other statin drugs might result in different outcomes remains to be investigated.

Future research will provide needed information about the optimal choice of drug, dose, and timing with regards to diagnosis and treatment for best results in cancer patients. Large, well-designed clinical trials are needed to clarify the potential role of statins in the context of cancer treatment.

Safety Considerations

Although statins are generally safe and usually cause minimal adverse events, some people experience negative side effects from statin therapy. The most common of these are muscle spasms, muscle pain, and new-onset diabetes.171 Because all statin drugs decrease production of coenzyme Q10 (CoQ10), long-term statin users benefit from CoQ10 supplementation.172-174

4 Aspirin

Background

Aspirin has been used for more than 100 years to treat pain, fever, and inflammation. Scientists have more recently begun to explore how aspirin might help people with cancer.175-177

Aspirin works by inhibiting a family of enzymes called cyclooxygenases (COXs). Cyclooxygenases help turn fatty acids into chemical messengers called prostaglandins and thromboxanes, which help promote pain signaling, blood clotting, and inflammation.178 Aspirin may fight cancer partly by reducing the inflammation that is critical to the development and growth of tumors.179,180 At the same time, by reducing platelet activity, aspirin may enhance anti-tumor immune activity and may have a role in supporting other anti-cancer immunotherapies.181,182 (For more information about aspirin in relation to immunotherapy, please see the "Cancer Immunotherapy" protocol.) Aspirin has also been shown to reduce cancer cell production of certain growth factors.183 In numerous preclinical studies, aspirin has demonstrated the ability to suppress a range of tumor cell activities and disrupt the tumor microenvironment in ways that interfere with tumor cell proliferation and metastasis.184 Because it exerts multiple effects on the biology of cancer cells, aspirin is considered to have pleiotropic anti-cancer properties.178

Observational (Correlational) Evidence

Numerous observational studies have linked low-dose aspirin use with reduced cancer incidence. A meta-analysis of 218 studies found regular low-dose aspirin use was correlated with lower overall cancer risk, as well as specific risks of gastric, esophageal, colorectal, pancreatic, ovarian, endometrial, breast, and prostate cancers.185 In addition, a large study performed in China that followed 204,170 people using low-dose aspirin and 408,339 age- and sex-matched non-aspirin users for about 10 years found low-dose aspirin use was associated with lower incidences of liver, stomach, colorectal, lung, pancreatic, and esophageal cancers, as well as leukemia; was not associated with incidences of multiple myeloma or cancers of the kidney, bladder, or prostate; and was associated with a slight increase in breast cancer incidence.186 However, an exploratory analysis of cancer outcomes in 10 studies from a meta-analysis of aspirin use and cardiovascular outcomes found no statistically significant effect of aspirin use on cancer incidence or mortality, compared with no aspirin use.187

Low-dose aspirin has also been correlated with better survival outcomes in multiple studies in people with cancer. In a study performed in the United States with 146,152 participants aged 65 years and older, regular low-dose aspirin use was linked to lower risks of cancer-related and all-cause death over 8.7–16.4 years of follow-up.188 Similarly, a study performed in France that included 111,025 participants aged 50–80 years found the 10-year risk of cancer and overall mortality were lower in low-dose aspirin users compared to non-users, with the protective association becoming stronger with each year of aspirin use.189 In a meta-analysis of 13 studies with a combined total of 65,768 cancer patients, low-dose aspirin use after cancer diagnosis was associated with lower mortality in patients with digestive tract cancers including colorectal, esophageal, and gastric cancers; however, an analysis of findings from four other studies that included 16,654 cancer patients failed to find an impact of low-dose aspirin use prior to cancer diagnosis.

Aspirin and gastrointestinal cancers: For many years, it appeared promising evidence was building for aspirin’s anti-cancer effects on gastrointestinal cancers. In 2016, accumulating research led the US Preventive Services Task Force (USPSTF) to recommend regular use of low-dose aspirin to prevent colorectal cancer in people 50–69 years with a low risk of bleeding. This was followed by similar recommendations by the US Multi-Society Task Force on Colorectal Cancer and the National Institute for Health and Care Excellence for patients with Lynch syndrome, a genetic disorder characterized by increased risk of colorectal cancer.190 Unfortunately, since then, large randomized controlled trials have not been able to demonstrate a protective effect for aspirin: one placebo-controlled clinical trial in 19,114 healthy older participants found long-term low-dose aspirin users had an increase in cancer-related and all-cause mortality rates.191 Another trial in 15,480 participants with type 2 diabetes reported no difference in risk of gastrointestinal cancer or any cancer with low-dose aspirin compared with placebo.192 In addition, in a randomized controlled trial in 2,536 Japanese patients with type 2 diabetes, low-dose aspirin did not impact cancer incidence.193 The USPSTF is in the process of updating its 2016 recommendations; although, the contents of the new recommendations have not been released as of mid-2020.

Aspirin and liver cancer: A meta-analysis of eight studies including data from more than 2.6 million participants found a strong association between aspirin use and lower incidence of hepatocellular carcinoma, the most common form of primary liver cancer. The analysis found aspirin users were 41% less likely to develop hepatocellular carcinoma than non-users during an average monitoring period of 8.9 years.194 Even in patients with high risk of liver cancer due to alcoholic cirrhosis or viral hepatitis, aspirin appears to reduce cancer risk.195-197 Aspirin use has also been reported to lower recurrence rate in liver cancer patients treated surgically.198

Aspirin and ovarian cancer: The Ovarian Cancer Cohort Consortium examined the findings from 12 observational studies with a total of 758,829 participants and found low-dose aspirin use six or more times per week was associated with a 10% reduction in ovarian cancer risk.199 A previous meta-analysis of 23 studies also found a correlation between regular use of low-dose aspirin and reduced incidence of ovarian cancer.200 An analysis of findings from the Nurses’ Health Study and Nurses’ Health Study II, which followed a total of 205,498 women, also found low-dose, but not regular-dose, aspirin was linked to lower ovarian cancer risk.201 Few studies have examined the relationship between aspirin use and survival in ovarian cancer patients. Some, but not all, evidence suggests aspirin use after ovarian cancer diagnosis may be linked to lower cancer-related mortality.202,203 In a small study that tracked outcomes in 77 women with clear cell ovarian cancer, an uncommon and typically aggressive form of ovarian cancer, aspirin use was found to be associated with lower mortality.204 Aspirin use before ovarian cancer diagnosis, on the other hand, does not appear to impact survival.202,205

Aspirin and gallbladder and biliary tract cancers: Two meta-analyses have found aspirin use may protect against the development of gallbladder cancer.206,207 Researchers have also reported better survival outcomes in biliary tract cancer patients who used aspirin regularly after their cancer diagnosis.208

Aspirin and gastric cancer: Two large meta-analyses of observational studies have found an association between long-term, regular, low-dose aspirin use and reduced risk of gastric cancer.209,210 The evidence for a protective effect comes from studies performed in China, Korea, and Sweden.211-214 Aspirin may have a particular role in protecting patients at increased risk due to their need for long-term treatment with proton-pump inhibitor (PPI) drugs to reduce gastric acid production. Aspirin’s effect on mortality in gastric cancer patients is unclear: a study performed in the United Kingdom found aspirin use was not associated with survival outcomes in those with gastric or esophageal cancers.215

Aspirin and breast cancer: Some evidence suggests low-dose, but not regular-dose, aspirin may lower the risk of breast cancer.216 A meta-analysis of 13 studies with a total of 857,831 participants found the use of low-dose aspirin two to seven times per week for more than five years was associated with a lower risk of breast cancer, and the relationship was stronger with more regular and longer duration of use.217 A study in 8,233 women whose risk of breast cancer was increased due to family history found regular aspirin users were less likely to develop breast cancer than non-aspirin users.218 However, one study in breast cancer survivors found no link between aspirin use and risk of a new cancer diagnosis involving the other breast.219 A systematic review of observational studies suggests low-dose aspirin during breast cancer treatment may also reduce metastases and cancer-related death.220 However, not all of the research is in agreement.221 Researchers are just beginning to explore the role of genetics and epigenetics in predicting whether individuals will benefit from aspirin in cancer prevention or treatment.222

Aspirin and prostate cancer: In a review and meta-analysis of data from observational studies, use of low-dose aspirin for three or more years was associated with reduced prostate cancer incidence.223 One study included men with PSA levels of 3 ng/mL or higher and monitored them for about 10 years. The study found men using low-dose aspirin had lower PSA levels at the beginning and end of the study; unexpectedly, overall mortality was higher in aspirin users compared with non-users.224 Other research has reported a link between daily low-dose aspirin use for cardiovascular protection and lower prostate cancer mortality.225,226 In one study, a protective association was only measurable after five years of aspirin use.227 But not all of the research is in agreement, and two meta-analyses of studies have failed to show a correlation between aspirin use and cancer-related or overall mortality in men with prostate cancer.228,229

Aspirin and pancreatic cancer: In two meta-analyses of observational studies, long-term, regular, low-dose aspirin use was associated with reduced risk of pancreatic cancer.230,231 One study performed in China found the risk in aspirin users was 46% lower than non-users.232 Nevertheless, not all studies have observed a relationship.108,233,234 For example, an analysis of data from 141,940 participants in the Nurses’ Health Study and the Health Professionals Follow-Up Study found no correlation between aspirin use and pancreatic cancer incidence; however, a possible protective effect in people with diabetes was noted.233

Aspirin and lung cancer: Findings from meta-analyses of observational studies regarding aspirin use and risk of lung cancer have been inconclusive.235,236 One large study performed in Korea that included data from nearly 1.3 million people found regular aspirin use was associated with a small reduction in risk of lung cancer.237 In a study performed in the United States, regular low-dose aspirin use was associated with lower lung cancer incidence only in men.238 Notably, in a study that included 19,894 people with COPD, a condition that raises the risk of lung cancer, aspirin use was correlated with a further increase in lung cancer risk.239 So far, studies in lung cancer patients have failed to find a relationship between low-dose aspirin use, before or after cancer diagnosis, and risk of cancer-related death.240-242

Aspirin and endometrial cancer: Although not all findings are in agreement, aspirin use has been found in multiple studies and meta-analyses to be correlated with reduced risk of endometrial cancer, particularly in overweight and obese women and with higher frequency and dosing of aspirin.243-247 There is conflicting evidence regarding the relationship between aspirin use and endometrial cancer survival.248,249

Aspirin and other cancers: A limited body of evidence also suggests aspirin use may be associated with a lower risk of brain cancer,250 and better outcomes in head and neck cancer patients,251-253 but may not be correlated with risk of thyroid cancer or survival in esophageal cancer patients.215,254

Causal Evidence - Clinical Trial Data

Few clinical trials of aspirin in cancer patients have been undertaken to date. The most recent trials have evaluated gastrointestinal cancer or overall cancer as endpoints, and results have not been promising.191-193

The ASPREE trial, a large clinical trial involving over 19,000 participants, first published in 2018, showed that 100 mg aspirin daily was associated with increased all-cause mortality in heathy older adults.255 In 2020, the authors of the ASPREE trial published an analysis of secondary endpoints covering cancer incidence and mortality. Among subjects randomized to aspirin therapy in ASPREE, there was no difference in overall cancer incidence or mortality compared with placebo. However, those taking aspirin were more likely to be diagnosed with metastatic cancer and to die from cancers that were diagnosed at stages III or IV than those taking placebo (although this remains a controversial finding and more research is still needed). The ASPREE authors suggested aspirin may have an adverse effect on later-stage cancers in older adults.256

In a trial that included 709 participants with a history of precancerous colon polyps, 300 mg aspirin daily for one year did not reduce polyp recurrence.257 At the time of this writing, the ADD-ASPIRIN placebo-controlled trial, which has enrolled 11,000 survivors of breast, prostate, colorectal, and gastroesophageal cancers, is underway in the United Kingdom, Republic of Ireland, and India to further test whether aspirin, at doses of 100 or 300 mg daily, can prevent cancer recurrence and increase chances of survival.258 Other randomized controlled trials in colorectal cancer patients are also underway.259-261

Safety Considerations

Although aspirin is available without a prescription, it is important to talk with a doctor before adding it to your treatment regimen. Because aspirin inhibits blood clot formation, daily aspirin use increases the risk of serious bleeding.262 It can also cause irritation and erosion of the stomach lining. People taking blood thinners, those who are at risk for gastrointestinal bleeding, and those with blood disorders may not be good candidates for daily aspirin use.

5 Diclofenac

Background

Diclofenac (Zipsor, Cataflam, Voltaren, and others) is a non-steroidal anti-inflammatory drug (NSAID). Oral, topical, and injectable preparations of diclofenac are used to treat inflammation, pain, and fever. Conditions such as arthritis, gout, migraine headache, and menstrual pain are often treated with diclofenac.263,264 In cancer care, diclofenac is used for post-operative and cancer-related pain and has an established role in treating actinic keratosis, a precancerous skin condition caused by chronic sun exposure.264

Diclofenac appears to interact with cellular pathways that regulate factors such as metabolism, equilibrium, pH, chemosensitivity, cell signaling, and inflammation.265 It is well known to inhibit COX enzymes, particularly COX-2, and thereby reduces production of prostaglandin E2 (PGE2) from arachidonic acid. PGE2 is a pro-inflammatory molecule that occurs in high levels in the tumor microenvironment and is involved in tumor initiation and progression.264,266 Diclofenac also decreases platelet function, which is important in the context of cancer because platelet activation can promote tumor growth through several mechanisms. Through these effects, diclofenac enhances anti-cancer immune function, reduces tumor-related new blood vessel formation, and increases cancer cells’ sensitivity to chemotherapy agents. Diclofenac has also been found to reduce glucose utilization, increase apoptosis, and suppress proliferation in cancer cells.264

Diclofenac has demonstrated anti-cancer effects in laboratory studies involving melanoma, leukemia, neuroblastoma, brain cancer, colorectal cancer, and prostate cancer cells.264,267,268 One study looked at the combined effects of metformin and diclofenac on brain cancer cells and found they had complementary cancer-killing effects on cellular metabolism.269 Other cell studies indicate diclofenac may work synergistically with the kinase inhibitor sorafenib (Nexavar) and platinum-based chemotherapy agent cisplatin (Platinol) to increase cancer cell death.270,271 Diclofenac has also demonstrated positive effects in animal models of colorectal, ovarian, brain, pancreatic, prostate, and oral cancers, as well as melanoma and neuroblastoma.264,272

Observational (Correlational) Evidence

Observational studies examining the link between diclofenac and cancer occurrence, recurrence, and mortality have had mixed findings.

Diclofenac and colorectal cancer: A study examining data from 2,308 patients undergoing surgery for colorectal cancer found those who took either ibuprofen (Advil) or diclofenac for at least two days around the time of surgery were 24% less likely to have a recurrence of their cancer compared with those who did not use any NSAIDs around surgery; however, NSAID use had no effect on five-year survival.273

Diclofenac and breast cancer: Diclofenac may lower breast cancer risk, but has not been found to lower risk of recurrence at distant sites in women treated for breast cancer. In 24,662 women with autoimmune disorders, those who reported using NSAIDs for at least three months during the previous nine years had a 63% lower incidence of breast cancer. Diclofenac, ibuprofen, piroxicam (Feldene), and selective COX-2 inhibitors (like celecoxib [Celebrex]) were each associated with decreased breast cancer risk, and even the women with low cumulative daily use appeared to be protected.274 A study that followed 34,188 breast cancer patients for a median period of 7.1 years after treatment found no relationship between use of NSAIDs post-diagnosis (with diclofenac being one of the most commonly used) and risk of breast cancer recurrence; however, NSAID use before diagnosis was associated with reduced rates of recurrence.275 Another study found use of diclofenac around the time of surgery to remove breast cancer had no effect on later development of distant metastases; however, use of another NSAID, ketorolac (Toradol), was correlated with decreased risk of metastatic recurrence, particularly in overweight and obese women.276

Causal Evidence - Clinical Trial Data

The only clinical trials investigating the role of diclofenac in cancer prevention have been in patients with precancerous actinic keratosis. COX activity is increased in skin cells exposed to UV light, raising levels of PGE2, and high PGE2 levels are associated with transformation to actinic keratosis and progression to squamous cell carcinoma of the skin.266 Multiple clinical trials and case reports show topical treatment with 3% diclofenac in a 2.5% hyaluronic acid solution produces high rates of response and remission and decreases long-term risk of progression to skin cancer in patients with actinic keratosis with few adverse effects.266,277 In two placebo-controlled trials, complete improvement was noted in 31–47% of participants treated with diclofenac/hyaluronic acid for 60 or 90 days, versus 8–19% of participants receiving placebo.278,279 Because of the strength of the evidence, topical 3% diclofenac/2.5% hyaluronic acid is FDA approved for use in actinic keratosis.266 However, compared with other available treatments, diclofenac may be less effective for achieving lasting remission.280,281

Although existing evidence suggests diclofenac may have anti-cancer properties related to its anti-inflammatory actions and other effects on cancer cell function, its potential benefits in the treatment of other types of cancer have yet to be explored in clinical trials.

Safety Considerations

Diclofenac can cause mild-to-moderate adverse side effects including abdominal pain, headache, dizziness, fluid retention, and a range of digestive symptoms. In rare cases, diclofenac can cause severe adverse effects such as gastrointestinal ulceration and bleeding, severe allergic reaction, anemia, pancreatitis, pneumonia, liver failure, and kidney damage and failure.263,264,282 Some evidence suggests diclofenac has greater toxicity to the heart and blood vessels than most other NSAIDs, and chronic use increases risk of heart attack, stroke, and cardiovascular death.282

6 Antihistamines

Background

Antihistamines interfere with interactions between histamine and either H1 or H2 histamine receptors. In general, H1 receptor blockers, like loratadine (Claritin), suppress histamine’s effect on the immune system and are used for treating allergic and anaphylactic reactions, while H2 receptor blockers, like cimetidine (Tagamet), inhibit gastric acid secretion and are used to treat gastrointestinal ulcers and other benign conditions of the stomach, esophagus, and duodenum. Both types of antihistamines are currently being studied for their potential utility in cancer care.

H1 Blocker: Loratadine

Many types of cancer cells express histamine receptors, through which histamine may increase cancer cell proliferation and tumor growth.283 The antihistamine loratadine, an H1 histamine receptor antagonist, has been found in preclinical research to induce cancer cell death by disabling cellular components called lysosomes (reservoirs of enzymes that break down intracellular molecules and dismantle the cell after cell death). Loratadine causes lysosomes to leak, releasing their cytotoxic contents into the cancer cell, and works synergistically with chemotherapy agents to cause cancer cell death.284 Loratadine has also been shown to damage cancer cell DNA, inhibit cancer cell proliferation, and enhance the cancer-killing effect of radiation and chemotherapy in preclinical research.285,286

Causal Evidence - Clinical Trial Data

A study performed in Denmark found the incidental use of either loratadine or astemizole (Hismanal) (a related antihistamine) by cancer patients was associated with reduced risk of death from any cause, and the association was stronger in those treated with chemotherapy.284 A similar 15-year study found ovarian cancer patients using loratadine or a related antihistamine were less likely to die from cancer.287 However, H1 blocker antihistamine use was not found to correlate with risk of new breast cancer in women who previously had cancer in the other breast.288

Loratadine may reduce some side effects of chemotherapy, increasing treatment tolerance. In women with ovarian cancer treated with paclitaxel (Taxol), those already using loratadine had a lower risk of developing treatment-related muscle and joint pain; furthermore, 75% of those who started loratadine after developing chemotherapy-induced joint and muscle pain had pain reduction. In a pilot trial, 600 women with breast cancer who were scheduled to undergo chemotherapy with pegfilgrastim (Neulasta) were randomly assigned to receive naproxen, loratadine, or no other treatment. Loratadine and naproxen both reduced chemotherapy-related bone pain, but loratadine caused fewer adverse side effects.289

Safety Considerations

Loratadine is generally well-tolerated but may cause headache or drowsiness in some individuals.290

H2 Blocker: Cimetidine

Cimetidine is an antacid medication that reduces acid production in the stomach by inhibiting the action of H2 histamine receptors. Cimetidine has demonstrated several effects that may interfere with cancer progression: inhibiting cancer cell proliferation; reducing T-regulatory cell function and thereby stimulating immune cell activity; preventing tumor growth by blocking histamine's action as a growth factor; and preventing the attachment of cancer cells to blood vessel walls.1,291 Cimetidine may also improve immune response to cancer by stimulating dendritic cell activity.292,293 Dendritic cells are immune cells with the critical job of activating other immune cells that then recognize and target invaders and infected and damaged cells, including cancer cells. For more information about dendritic cells, please see the "Cancer Immunotherapy" protocol.

Causal Evidence - Clinical Trial Data

In a study that evaluated a combined treatment approach, cimetidine was administered alongside other drugs that inhibit an enzyme involved in a type of brain tumor known as glioblastoma. The enzyme, called glycogen synthase kinase-3 beta (GSK-3β), makes brain tumor cells more aggressive. The combination of cimetidine, lithium (Eskalith, a mood stabilizer), olanzapine (Zyprexa, an antipsychotic medication), and valproate (Depakote, an anti-seizure medication) appeared to improve survival in patients with brain tumor that had active GSK-3β.294

Several trials have investigated the effect of cimetidine in colon cancer treatment. One study reported on outcomes in 38 colon cancer patients who did or did not receive cimetidine therapy, along with chemotherapy, around the time of cancer surgery; the addition of cimetidine was associated with longer time to cancer recurrence and longer cancer survival.295 A meta-analysis of five randomized controlled trials with a combined total of 421 participants found cimetidine, as an adjunct to surgery, prolonged survival in colorectal cancer patients.

Although findings have been mixed, researchers continue to investigate the possible role of cimetidine in the treatment of kidney cancer. In one promising trial, cimetidine was combined with a COX-2 inhibitor (meloxicam [Mobic]) and either an ACE inhibitor (perindopril [Aceon]) or angiotensin II receptor blocker (candesartan [Atacand]) and used adjunctively with the chemotherapy agent interferon in 51 patients with advanced kidney cancer. Four patients achieved a complete response, seven achieved a partial response, and the remaining 24 had no disease progression for at least six months. Another pilot trial investigated cimetidine, at a dose of 600 mg four times daily, as the only treatment in 42 patients with advanced kidney cancer; two participants had complete remission lasting more than two years.

Adjuvant treatment with cimetidine has been noted to prolong survival in gastric cancer patients, and this effect may be more pronounced in individuals with more advanced cancer.296,297 However, in one controlled trial that included 442 gastric cancer patients, the addition of cimetidine to treatment had no effect on survival.298

Cimetidine treatment may be particularly effective in patients whose tumor cells have high numbers of surface proteins called Lewis A and Lewis X antigens. These antigens are seen particularly in some breast and pancreatic cancers, as well as about 70% of colon cancers. Cimetidine is thought to inhibit interactions between Lewis A and Lewis X antigens and blood vessel walls that can lead to tumor cell attachment and invasion.299,300 One clinical trial in gastric cancer patients found that, in a subgroup of participants expressing Lewis X antigens, the 10-year survival rate was 95.5% in those receiving cimetidine and 35.1% in those not receiving cimetidine; in participants with Lewis A antigens, 10-year survival was 90.9% with cimetidine and 20.1% without cimetidine.301

Safety Considerations

Although cimetidine is available without a prescription, it is important to talk with a doctor before adding cimetidine to your treatment program. Cimetidine can interact with several commonly prescribed medications, such as digoxin (Lanoxin), theophylline (Theobid or Theo-Dur), phenytoin (Dilantin), warfarin (Coumadin), lidocaine (Xylocaine), propranolol, and antidepressants. These drug interactions can result in increased risk of side effects and altered efficacy of the medications.302 Because of cimetidine’s potential for many drug-drug interactions, other H2 blockers are now preferred in the general clinical setting. However, other H2 blockers have generally not been studied in the context of cancer to the extent cimetidine has.

7 Beta-Blockers

Background

Beta-blockers are a class of medications that inhibit a type of adrenaline receptor known as beta-adrenergic receptors. These receptors are activated by neurohormones called catecholamines, particularly epinephrine and norepinephrine. Two subtypes of beta-adrenergic receptors, β1 and β2, are found throughout the body, and different drugs within the beta-blocker family have different effects. Non-selective beta-blockers, such as propranolol (Inderal), inhibit both β1 and β2 receptors; selective beta-blockers, like atenolol (Tenormin) and metoprolol (Lopressor), inhibit only β1 receptors. Beta-blockers have a long history of use in treating hypertension, but certain drugs in this class are also used to prevent migraine headaches, reduce anxiety, and treat essential tremor. Most recently, beta-blockers have attracted interest for their potential anti-cancer effects.303

Levels of norepinephrine and epinephrine rise in response to stress, and cancer diagnosis and treatment are often accompanied by intense psychological and physical stress and loss of quality of life. Beta-adrenergic receptors are found on cancer and immune cells, allowing norepinephrine and epinephrine to influence cancer progression by enhancing tumor-related inflammatory signaling, tumor cell invasion and migration, and formation of new blood and lymph vessels, while disrupting anti-cancer immune activity.303-305 Decreasing the cancer-promoting effects of these stress-response neurohormones has the potential to improve cancer outcomes.306 Preclinical evidence suggests beta-blockers may alter the tumor microenvironment in ways that inhibit cancer growth and spread.303,304 Furthermore, beta-blockers have been found to reduce cardiac toxicity from anthracyclines, a widely used class of chemotherapy drugs.307

Observational (Correlational) Evidence

The potential anti-cancer benefits of beta-blockers were first identified when correlations were reported between incidental propranolol use and reduced risk of many types of cancer. Non-selective beta-blocker use has since been associated with reduced cancer recurrence in melanoma, ovarian cancer, and breast cancer patients, and increased cancer-related survival in patients with melanoma and ovarian, pancreatic, colorectal, liver, and breast cancers.303,308-310 On the other hand, studies in patients with lung,311,312 prostate,311,312 head and neck,313,314 and endometrial315 cancers have reported no benefit from beta-blocker use as of early 2021. An observational study that used data from 30,020 subjects with breast, colorectal, or lung cancers did not find a survival advantage in those who were incidentally taking beta-blockers around the time of surgery to remove their tumors.316

Studies in women with ovarian cancer have had mixed findings. In one observational study, women with ovarian cancer who used non-selective beta-blockers during the year following their cancer diagnosis were more likely to survive their cancer than women taking no blood pressure-lowering medications.317 Another study involving ovarian cancer patients found certain factors were associated with a benefit from beta-blocker use; specifically, in those over 60 years old, those with existing cardiovascular or other chronic disease, and those who used beta-blockers for one year or longer, beta-blockers were linked to increased survival during at least three years of monitoring.318 However, a large study that followed 6,197 ovarian cancer patients found both selective and non-selective beta-blocker use during the five years after cancer diagnosis were associated with lower rates of cancer-related and overall survival.319

Propranolol and other beta-blockers may be helpful in cancers with few treatment options and poor prognoses. One study found propranolol use was correlated with improved survival in liver cancer patients with metastatic disease, but not those whose tumors were amenable to surgery.320 In colorectal cancer patients requiring emergency surgery, those who had been taking beta-blockers prior to surgery had better short-term (30-day) and long-term (one-year) survival.321 In women with triple-negative breast cancer, a type of breast cancer that is difficult to treat and particularly aggressive, beta-blocker use was also linked to better outcomes. However, the link between cancer outcomes and beta-blocker use is challenging to interpret. Adequately controlled hypertension has been associated with better cancer outcomes, so the apparent benefits could be related to better blood pressure control in general.322

Causal Evidence - Clinical Trial Data

A small randomized controlled trial published in June 2020 found propranolol plus the COX-2 inhibitor etodolac (Lodine) improved tumor molecular markers of malignant and metastatic potential and non-significantly reduced recurrence rates among individuals who underwent surgery for colorectal cancer. The propranolol and etodolac were administered for 20 days perioperatively, starting five days before surgery. Although the trial was small and not initially designed to assess differences in survival, the trends toward increased survival in the recipients of propranolol and etodolac warrant larger trials sufficiently powered to assess clinical outcomes in colorectal cancer patients.323

Some evidence suggests beta-blockers may have a role as an adjunctive therapy for breast cancer.324,325 In a randomized placebo-controlled trial, 60 women with early-stage breast cancer received either propranolol or placebo for seven days prior to surgical tumor removal. Cells from tumors of women who received propranolol were found to express fewer markers associated with cancer progression than cells from tumors of women who received placebo.321

In a small randomized controlled trial that enrolled 25 patients undergoing hematopoietic cell transplantation (HCT) for multiple myeloma, propranolol administered before and after the transplant led to changes in cellular and molecular pathways that are generally associated with favorable outcomes. In this study, 13 subjects received placebo and 12 subjects received propranolol starting seven days before HCT. The starting dose of propranolol was 20 mg twice daily, which was increased to 40 mg twice daily after one week if there were no adverse effects observed on the 20 mg dose. Propranolol was continued until 28 days after HCT. After analyzing the data, the authors concluded that, “Peri-HCT propranolol inhibits cellular and molecular pathways associated with adverse outcomes. Changes in these pathways make propranolol a potential candidate for adjunctive therapy in cancer-related HCT.”326

The body of research on beta-blockers and cancer outcomes is still in an early stage. Future research will help elucidate the anti-cancer potential of specific beta-blockers and how type of cancer, timing of beta-blocker use, and use of other therapies (surgery, radiation, chemotherapy) influence the effects.310,327

Safety Considerations

Propranolol and other beta-blockers have a long history of safe use. Typical side effects, such as fatigue, insomnia, cold extremities, and less commonly digestive symptoms, are mild. These effects can often be avoided by starting with a low dose and building slowly to a therapeutic dose. Discontinuing beta-blockers must also be done slowly. Beta-blockers are contraindicated in people with low blood pressure, asthma, uncontrolled heart failure, metabolic acidosis, severe peripheral artery disease, and cardiogenic shock.328

8 Valproic Acid

Background

Valproic acid, a short-chain fatty acid with anticonvulsant activity, is used to treat epilepsy, as well as bipolar disorder and migraines. Preclinical studies indicate valproic acid inhibits cancer by inducing normal cell maturation and apoptosis. Its apparent anti-cancer effects are likely to be due in part to its ability to affect epigenetic regulation of gene expression by inhibiting enzymes involved in gene activation.329,330

Valproic acid has been found to increase sensitivity to chemotherapy agents in ovarian, pancreatic, and breast cancer cells, and increase radiation sensitivity in breast cancer cells cultured in the laboratory.331-334 In liver cancer cells, valproic acid was found to suppress tumor cell signaling and induce expression of receptors that could be targets of other anti-cancer drugs.335 In prostate cancer cells, valproic acid inhibited cell proliferation, migration, and invasion, and its effects were enhanced by combining it with other anti-cancer agents.336

Causal Evidence - Clinical Trial Data

In preliminary clinical research, valproic acid has been found to have synergistic effects with the chemotherapy drug combination 5-azacytidine (Vidaza) plus all-trans retinoic acid in patients with blood cancers (myelodysplastic syndromes and acute myeloid leukemia),337 and with 5-fluorouracil (5-FU, Adrucil) in pancreatic cancer patients.338 In a pilot trial, treatment with valproic acid led to disease stabilization for one year in four of eight individuals with low-grade carcinoid or pancreatic tumors.339 In another pilot trial that included 32 patients with advanced solid tumors that were unresponsive to standard therapies, valproic acid combined with two chemotherapy drugs (azacitidine and carboplatin [Paraplatin]) led to minor responsiveness or disease stabilization lasting four months or longer in six participants.340 As of late-2020, a trial is ongoing to test valproic acid in combination with bevacizumab (Avastin) and oxaliplatin/fluoropyrimidine regimens in people with RAS-mutated metastatic colorectal cancer341; the estimated completion date for this trial is August 2022.

Not all available evidence supports the use of valproic acid as a cancer adjunct therapy. Valproic acid was tested in combination with paclitaxel in 66 individuals with advanced gastric cancer. Outcomes were no different in the group receiving paclitaxel plus valproic acid than in the paclitaxel-only group.342

Safety Considerations

Valproic acid’s usefulness can be limited by adverse side effects such as fatigue and low white blood cell numbers, as well as neurological symptoms such as disorientation, memory loss, lethargy, and loss of muscular control.340,343 Valproic acid has also been linked to liver failure, and liver function tests should be monitored regularly during use. Valproic acid may also cause serious pancreatic damage. It is contraindicated in women who are pregnant or may become pregnant.

9 Dichloroacetate

Background

Dichloroacetate, a drug used to treat lactic acidosis, genetic conditions affecting mitochondrial function, and diabetes, has demonstrated anti-cancer effects in laboratory and animal studies. In animal research, dichloroacetate has been shown to decrease tumor size, slow tumor cell proliferation, and inhibit metastasis.344

Dichloroacetate appears to work by interfering with cancer cell metabolism. Cancer cells disproportionately use glycolysis to metabolize glucose, which leads to accumulation of lactic acid in the tumor microenvironment. Lactic acid stimulates tumor growth and inhibits apoptosis.344-346 Dichloroacetate shifts metabolism away from glycolysis toward mitochondrial energy production and reduces lactic acid accumulation, altering the tumor microenvironment away from one that supports cancer growth and spread. Furthermore, by promoting mitochondrial metabolism of glucose, dichloroacetate induces cellular changes that favor normal apoptotic processes.344,347

Multiple preclinical studies have found dichloroacetate can increase sensitivity and reduce resistance to chemotherapy drugs and radiation therapy in certain cancer types, and may enhance the actions of other potential anti-cancer agents such as propranolol and metformin.344,348 Importantly, dichloroacetate may even sensitize cancer stem cells—which are thought to be central to the development of resistance and cancer recurrence—to anti-cancer treatment.349,350

Observational (Correlational) Evidence

Several case studies using dichloroacetate in cancer patients have been published. One case report describes a 32-year-old man with malignant melanoma who experienced remission and disease stabilization for more than four years after adding dichloroacetate to a naturopathic treatment program. Similarly, in a 57-year-old woman with stage IV colon cancer, dichloroacetate therapy in combination with chemotherapy, bevacizumab, and metformin resulted in stabilization of disease for almost four years. Positive responses to dichloroacetate therapy have also been reported in patients with recurrent cancers that were unresponsive to standard cancer care.351,352

Causal Evidence - Clinical Trial Data

In a pilot trial, seven participants with multiple myeloma were treated with dichloroacetate; one had a full response and two had partial responses to treatment after one month. The researchers also noted specific genetic markers related to drug metabolism and efficacy.353

Safety Considerations

The usefulness of dichloroacetate is limited mainly by its neurotoxicity. Neurological side effects from dichloroacetate resolve after it is discontinued.344,354

10 Low-Dose Naltrexone

Background

Naltrexone is an opioid antagonist used to treat alcohol and opioid dependence. However, at low doses (roughly 1/10th of the typical addiction treatment dosage, commonly 4.5 mg/day), naltrexone has been used for off-label treatment of a variety of pain-related conditions, including fibromyalgia, multiple sclerosis, and Crohn’s disease.355-357 At this dosage level, naltrexone also exhibits immunomodulatory effects and has shown early promise for the treatment of autoimmune disorders and cancer.358

Naltrexone has high affinity for μ-opioid receptors, which are overexpressed in a number of different cancers.359-363 In healthy cells, interactions between opioid growth factor (OGF) and opioid growth factor receptor (OGFr) help regulate DNA synthesis and cell renewal. In fact, OGF has been shown to depress tumor proliferation and possess broad immune modulating properties, with important implications for regulating the tumor microenvironment and promoting anti-tumor immunity.364 Targeting the OGF-OGFr axis with naltrexone is being actively investigated in preclinical models.365,366 Naltrexone has also been found to bind toll-like receptor 4, disrupting cellular pro-inflammatory responses.367-369 In a mouse colorectal cancer xenotransplant model, low-dose naltrexone increased macrophage activity and expression of pro-apoptotic factors, suggesting a multi-faceted immunoregulatory function.370,371 In vitro, low-dose naltrexone enhanced the cell-killing activity of the chemotherapy drug oxaliplatin in colorectal cancer cells, whereas higher-dose naltrexone was not as effective.371

Low-dose naltrexone has been used successfully in multiple animal models of carcinomas. In a mouse model of solid Ehrlich carcinoma, treatment with low-dose naltrexone resulted in reduced tumor weight and volume. In combination with the chemotherapeutic drug 5-FU, low-dose naltrexone was also associated with reduced expression of an important antiapoptotic factor in treated mice.372 Similar results were seen in a mouse model of ovarian cancer, in which low-dose naltrexone in combination with the chemotherapeutic drug cisplatin reduced DNA synthesis and inhibited tumor progression.373 In a study of 60 dogs with mammary carcinomas, the addition of low-dose naltrexone to chemotherapy was associated with prolonged survival, fewer chemotherapy-related adverse events, and better quality of life.374

Observational (Correlational) Evidence

Studies examining the efficacy of low-dose naltrexone as an anti-cancer agent in humans have been lacking, and current evidence for its clinical use is based on several case reports. The earliest of these reports described the use of low-dose naltrexone in combination with alpha-lipoic acid in four patients with metastatic and non-metastatic pancreatic cancer. As early as four and five months after beginning the combination treatment, two of the patients demonstrated no evidence of disease on position emission tomography (PET) scan, and no appreciable progression of malignancy was reported up to 78 months after initial presentation.375,376 Use of a similar protocol was also described in a 64-year-old man diagnosed with metastatic renal cell carcinoma, which resulted in stable disease and disappearance of radiological evidence of malignancy for up to nine years after diagnosis.377 Treatment with low-dose naltrexone has also been described for a 50-year-old man with a history of prostate and lung cancer. The patient began low-dose naltrexone treatment after halting standard chemotherapy due to intolerable side effects and exhibited no signs of disease recurrence almost four years after initiating treatment. This patient has been maintained throughout on an array of 11 other medications for other chronic conditions.378 Successful use of low-dose naltrexone treatment has also been reported in two cases of hepatoblastoma, in an infant and a young child. At the time of the report, the children had disease-free survival of 10 and five years, respectively.379

Safety Considerations

The preliminary reports described above also suggest low-dose naltrexone exhibits a favorable safety profile as an anti-cancer agent and is associated with minimal side effects.375-379 A systematic review of randomized controlled trials of naltrexone use in various conditions, compared with placebo, found that naltrexone does not increase the risk for serious adverse events, providing a firm basis for researchers to further investigate low-dose naltrexone’s therapeutic potential.380

11 PDE-5 Inhibitors

Background

Phosphodiesterases (PDEs) are a family of cellular enzymes that catalyze the breakdown of two cyclic nucleotide compounds: cyclic adenosine monophosphate (cAMP) and cyclic guanine monophosphate (cGMP). For this reason, PDEs are also called cyclic nucleotide phosphodiesterases. cAMP and cGMP are important second messengers—a specific type of signaling molecule—that regulate the transmission of signals within cells and in this way modulate such fundamental cellular activities as cell division, apoptosis, metabolism, inflammation, and gene expression.381,382 Nearly 100 distinct PDEs have been identified, and these are broadly categorized according to their ability to break down cAMP, cGMP, or both. The PDE-5 category targets cGMP.381,382

PDE-5 inhibitors are used to treat erectile dysfunction, pulmonary hypertension, and benign prostatic hyperplasia. They are also under investigation as possible agents for treating diabetes, atherosclerosis, metabolic syndrome, heart failure, and stroke, as well as lower urinary tract symptoms.381-383 Three PDE-5 inhibitors are currently of particular interest for cancer therapy:

  • Sildenafil (Viagra)
  • Tadalafil (Cialis)
  • Vardenafil (Levitra)

Impaired production of cyclic nucleotides or overexpression of PDEs may contribute to cancer cell growth and migration, and targeted PDE inhibition may promote tumor cell apoptosis and decrease uncontrolled tumor cell growth.382,383 High levels of PDE-5 are reported to be present in several types of cancer cells, including breast, prostate, lung, and colorectal cancers, as well as melanoma.383

PDE-5 inhibition has been found to trigger both cancer cell apoptosis and anti-cancer immune activation. PDE-5 inhibitors, mainly in combination with various chemotherapy drugs, have been shown to enhance suppression of cancer cell activity and increase apoptosis in cancer cells or other preclinical models of cancers such as lung, breast, prostate, colorectal, bladder, liver, thyroid, head and neck, and brain cancers, as well as several blood cancers.383,384

Observational (Correlational) Evidence

The incidental use of PDE-5 inhibitors, usually for erectile dysfunction, may be associated with lower risk of some cancers as well as better outcomes in certain cancers. There is evidence for a benefit in colorectal cancer; their effects on prostate cancer risk is less clear. However, observational studies have found an association between the use of PDE-5 inhibitors for erectile dysfunction, and a higher risk of malignant melanoma and possibly other skin cancers.385-387 Although plausible mechanisms by which PDE-5 inhibition could promote melanoma have been described, multiple authors suggest this relationship is likely a result of confounding and thus non-causal.384,388,389

PDE-5 inhibitors and colorectal cancer: In a study that included more than 221,000 men, having used a PDE-5 inhibitor for erectile dysfunction was associated with an 18% lower risk of colorectal cancer compared with never having used one.390 In a study that examined data from 36,020 men diagnosed with benign colorectal polyps during a 10-year period, those taking PDE-5 inhibitors were found to have a 35% reduced incidence of colorectal cancer. Greater cumulative PDE-5 inhibitor doses were associated with greater protection.391 However, another study in 200,000 male participants whose data were collected for The Health Improvement Network found no association between PDE-5 inhibitor use and colorectal cancer incidence.392

A 2020 study investigated the relationship between PDE-5 inhibitor use and colorectal cancer outcomes. The study, which included 12,465 male colorectal cancer patients, found those who used PDE-5 inhibitors after their cancer diagnosis had an 18% lower chance of colorectal cancer-specific death and a 15% lower risk of distant metastasis during 4.25 years of monitoring. These associations were stronger in those who had undergone open surgery to remove their tumor.393

PDE-5 inhibitors and prostate cancer: A study that followed 6,501 men for four years found PDE-5 inhibitor use was not correlated with prostate cancer incidence overall, but was associated with a 33% risk reduction in North American participants; however, this risk reduction was not statistically significant.394 A systematic review and meta-analysis found no link between PDE-5 inhibitor use and prostate cancer risk or risk of recurrence in prostate cancer patients.395,396 Although one study found that PDE-5 inhibitor use increased the risk of biochemical recurrence (a rise in PSA level) in men treated surgically for prostate cancer,397 multiple other studies and a meta-analysis have failed to confirm that increased risk.398-402

PDE-5 inhibitors and other cancers - case reports: The use of sildenafil for erectile dysfunction was reported to improve the clinical course in five cases of men with a rare incurable blood cancer called Waldenström’s macroglobulinemia. In one case, complete remission occurred after starting sildenafil.403 In another case series, sildenafil use was associated with long-term disease-free survival in two of three cases of penile cancer, which was attributed to possible sensitization to radiation treatment by the drug.404,405 A case report described how the addition of tadalafil led to enhanced treatment response in a patient with relapsed multiple myeloma that had previously been unresponsive to treatment. The researchers suggested tadalafil likely induced an anti-myeloma immune response by reducing the function of myeloid-derived suppressor cells, which suppress anti-tumor immunity and promote tumor progression.405,406

Causal Evidence - Clinical Trial Data

The effect of tadalafil treatment, at gradually diminishing doses starting at 40 mg and eventually lowered to 5 mg, on melanoma was investigated in an open trial that included 12 patients with metastatic melanoma, 11 of whom had already received standard cancer treatment and all of whom had evidence of cancer progression. Tadalafil was associated with increased tumor targeting by immune cells, and three participants experienced stabilization of their condition. The tadalafil dose did not correlate with clinical response in this study.407 Tadalafil has similarly been found to reduce tumor-related immune suppression in patients with head and neck cancer in two placebo-controlled trials.408,409 A number of clinical trials that include tadalafil in patients with various types of cancer are underway as of mid-2020.410

Safety Considerations

Adverse side effects such as nausea and vomiting, headache, back pain, fatigue, and weight loss have been reported in men using PDE-5 inhibitors.407,409 Other known side effects include dizziness, visual disturbance, red eyes, heart palpitations, rapid heartbeat, high or low blood pressure, and nose bleeds. In rare cases, PDE-5 inhibitors have been linked to cardiovascular events, retinal vascular occlusion (blood clot in a tiny vessel in the eye), and sudden hearing loss.384

12 Chloroquine and Hydroxychloroquine

Background

Chloroquine (CQ, Avloclor) has historically been used to prevent and treat malaria, and its safer metabolite, hydroxychloroquine (HCQ, Plaquenil), has more recently been used to treat autoimmune diseases such as systemic lupus erythematosus, discoid lupus, Sjögren’s syndrome, and rheumatoid arthritis.411,412 These medications inhibit an important cellular process called autophagy, through which cells clean up and recycle misfolded proteins, damaged organelles, and other degraded components as a strategy to adapt to stressful conditions.413 Autophagy functions as a tumor suppressor, preventing the initiation of cancer. However, autophagy could support cancer progression by contributing to the maintenance of cancer stem cells, helping tumors survive cellular stresses, and stimulating resistance to chemotherapeutic agents.413-415 This dual effect complicates the use of autophagy-inhibiting medications in cancer therapy.413 In addition to inhibiting autophagy, some evidence suggests CQ and HCQ may suppress cancer growth and activity by inhibiting certain signaling pathways in cancer cells, altering the tumor microenvironment, and interfering with tumor blood vessel supply.411 Importantly, while autophagy is overactive in many tumor cells, only certain types are sensitive to autophagy inhibitors, and these considerations are important to clinical practice.412

CQ and HCQ have been found to suppress tumor initiation and growth in numerous cell culture and animal models of various types of cancer, including brain, liver, breast, pancreatic, and colorectal cancers, as well as lymphoma and melanoma. On the other hand, some studies have shown CQ and HCQ can promote cancer growth and metastasis of some types of cancer, indicating the importance of identifying the tumor characteristics associated with a likely benefit from CQ or HCQ. One proposed indicator of potential HCQ/CQ efficacy is p53 status (p53 is an important tumor-suppressor protein). Another is the degree to which different tumor cells and tumor types are dependent on autophagy. Dose timing relative to chemotherapy may be important as well, with some authors hypothesizing that CQ should be administered after chemotherapy, not before.411

Multiple preclinical studies have noted CQ and HCQ can enhance the efficacy of other cancer treatments by increasing cancer cell sensitivity to therapeutic agents. In fact, CQ and HCQ have been tested in combination with more than 40 anti-cancer therapies, including chemotherapy drugs, radiation therapy, monoclonal antibodies, hormone therapies, and tyrosine kinase inhibitors, and have led to decreased tumor growth and improved survival in some cases.411,413 However, some of this work has been criticized, as the positive results were obtained primarily in immune-deficient animals, while some results were disappointing in immune-competent hosts.413

Causal Evidence - Clinical Trial Data

Chloroquine: Clinical Evidence

CQ and brain cancer: A randomized controlled trial in 18 patients with glioblastoma multiforme found survival time was significantly increased in those whose post-surgical treatment with radiation and chemotherapy was augmented by 24 to 50 months of CQ therapy.416 A similar trial by the same research group found CQ therapy for 12 months after surgery markedly prolonged survival time relative to placebo in 30 glioblastoma multiforme patients under age 60 who were treated with chemotherapy and radiation after surgery.417 This research group published a retrospective report describing significantly increased survival time in 41 glioblastoma multiforme patients who received CQ in addition to standard treatment compared with 82 patients who did not.418

Several case reports and some preclinical research indicate the addition of CQ may restore sensitivity to the targeted cancer drug vemurafenib (Zelboraf) in brain cancer patients who developed resistance to the drug.419,420

Two clinical trials have examined the effect of CQ in patients with brain metastases (secondary brain tumors). In one placebo-controlled trial, 39 cancer patients with brain metastases received whole brain irradiation plus CQ or placebo for four weeks; although the CQ group did not have a higher response rate or overall survival, they did have longer progression-free survival.421 An uncontrolled trial found CQ in combination with whole brain radiation therapy for three months led to a complete response in two of 16 patients with brain metastases, partial response in 13, and tumor stabilization in one.422

CQ and multiple myeloma: Two clinical trials evaluated CQ as add-on therapy in multiple myeloma patients. In an uncontrolled trial, eight evaluable participants with relapsed and unresponsive multiple myeloma had CQ added to chemotherapy with bortezomib (Velcade) plus cyclophosphamide (Cytoxan). Three subjects had a partial response and one had tumor stabilization, while the other four progressed.423 A controlled trial in 38 multiple myeloma patients, however, found adding CQ to treatment with cyclophosphamide plus prednisone had no impact on treatment response.424

CQ and pancreatic cancer: In an uncontrolled trial in nine patients with metastatic or non-surgically treatable pancreatic cancer, three participants had a partial response and two had tumor stabilization following treatment with the chemotherapy drug gemcitabine (Gemzar) plus CQ.425

Hydroxychloroquine: Clinical Evidence

HCQ and solid tumors: In 25 stage IV cancer patients with various solid tumors who had not responded to standard treatment, the addition of rapamycin plus HCQ to treatment for at least three months led to partial response in 10 participants and tumor stabilization in 11 participants. In all, 84% of evaluable participants showed a positive clinical effect.426 Another trial in 27 patients with solid tumors was designed as a safety, tolerability, and pharmacology study of HCQ plus the chemotherapy agent vorinostat (Zolinza). The authors reported a partial response in one patient with kidney cancer and prolonged tumor stabilization in two patients with colorectal cancer.427 A pilot trial tested a combination of HCQ and the investigational drug MK-2206 in 35 patients with advanced solid tumors, but found minimal evidence of anti-tumor activity and a substantial number of adverse side effects.428

HCQ and pancreatic cancer: In a clinical trial in 20 previously treated metastatic pancreatic cancer patients, HCQ as a stand-alone therapy had little therapeutic efficacy, was associated with serious adverse side effects in two participants, and did not reliably inhibit autophagy.429 Another trial in 35 pancreatic cancer patients deemed unlikely to benefit from surgery tested the combination of gemcitabine with HCQ for 31 days prior to surgery. Nineteen participants had reductions in levels of a cancer biomarker, and 29 proceeded to surgery. Among surgically treated participants, 77% had a successful surgical outcome with clean margins, and those who had a significant increase in an autophagy marker in response to HCQ had significantly longer disease-free and overall survival. According to the study’s authors, gemcitabine plus HCQ resulted in better outcomes than historical treatments in similar patients.430 Another randomized controlled trial included 64 pancreatic cancer patients and compared treatment with gemcitabine and paclitaxel to the same chemotherapy regime plus HCQ for two cycles, followed by surgery. Those who received HCQ had better responses than those who did not; however, there were no differences in overall survival or time to relapse.431 An open-label trial with 112 participants also found no survival benefit from adding HCQ to pancreatic cancer treatment with gemcitabine and paclitaxel after one year, although the overall response rate was significantly greater in the HCQ group. Severe adverse effects were considerably greater in the HCQ arm.432

HCQ and colorectal cancer: In 20 participants with metastatic colorectal cancer that was unresponsive to treatment, HCQ plus vorinostat appeared to enhance anti-tumor immune activity. Although no partial or complete treatment responses were noted, five of the participants had stable disease for more than 16 weeks.433

HCQ and multiple myeloma: A safety and preliminary efficacy trial in 21 participants with relapsed or unresponsive multiple myeloma found that two weeks of HCQ alone, followed by the addition of bortezomib for a median of 14 weeks (range: 1‒77 weeks), led to very good partial response in three patients, minor response in three, periods of no progression in 10, and immediate progression (no treatment response) in six.434

HCQ and sarcoma: An uncontrolled trial enrolled 10 patients with soft tissue sarcoma that had not responded to standard treatment. After two weeks of therapy with a combination of sirolimus (Rapamune, also known as rapamycin) and HCQ, six participants had partial response, three had cancer stabilization, and one had cancer progression. Although tumor cells’ use of glucose for energy production was reduced with treatment, none of the tumors decreased in size. Nausea, diarrhea, constipation, and skin rash were reported as mild side effects of treatment.435

HCQ and melanoma: In a trial designed to study the safety, tolerability, and pharmacology of a combination of HCQ plus temozolomide in 40 patients with solid tumors, 27 of whom had malignant melanoma, treatment resulted in partial response or cancer stabilization in 38% of those with melanoma.436 A similar trial, performed by the same research group, in patients with advanced solid tumors and melanoma found cancer stabilization in 14 of 19 melanoma patients treated with temsirolimus (Torisel, a derivative of sirolimus) plus HCQ. However, no partial or complete responses were noted.437

HCQ and lung cancer: In a safety, tolerability, and pharmacology study in 27 patients with advanced non-small cell lung cancer treated with HCQ alone or in combination with the chemotherapy agent erlotinib (Tarceva), only one patient receiving combination therapy experienced a partial response.438 In another uncontrolled trial in 40 untreated metastatic non-small cell lung cancer patients given four to six cycles of standard chemotherapy augmented by HCQ, 10 patients had some treatment response and six others had cancer stabilization.439

HCQ and kidney cancer: In patients with advanced clear cell renal cell carcinoma who had undergone at least one prior anti-cancer protocol, treatment with HCQ plus everolimus (Afinitor, Zortress) resulted in partial response in two of 33 participants, stabilization in 20, and progression-free survival for six months in 15.440

Safety Considerations

Multiple clinical trials have been initiated to study the safety of CQ and HCQ in cancer treatment, mainly in combination with other anti-cancer therapies, and to identify tolerable doses. It is important to note that few of these trials were designed to evaluate the efficacy of CQ and HCQ in cancer treatment. CQ and HCQ, though tolerable for most patients in these studies, were associated with several adverse side effects, including fatigue, loss of appetite, and digestive symptoms. Higher and longer-duration doses in particular appeared to cause more serious adverse effects.441 In addition, serious side effects such as heart rhythm abnormalities, hypoglycemia, bone marrow toxicity leading to decreased blood cell production, heart muscle damage, and irreversible damage to eye tissue causing visual impairment have been reported in some patients treated with CQ or HCQ.411,442,443

13 Clarithromycin

Background

Clarithromycin (Biaxin) is a member of the class of antibiotics known as macrolides. It is widely used as part of therapy to eradicate gastric H. pylori infection, and is also used to treat other bacterial infections, as well as Lyme disease. Because H. pylori infection increases the risk of gastric cancer, clarithromycin can play a role in gastric cancer prevention.444,445 In addition, a growing body of evidence suggests it may help treat other types of cancer, including lymphoma, multiple myeloma, chronic myeloid leukemia, and non-small cell lung cancer.446

Studies in cancer cells and animals show clarithromycin slows tumor growth, induces tumor cell apoptosis, and inhibits tumor-associated formation of new blood vessels in models of cancers such as melanoma, lymphoma, lung, and colorectal cancers.446,447 Clarithromycin has also been found to enhance the anti-cancer effects of some chemotherapy agents and modulate levels of inflammatory cytokines related to cancer progression in animals with melanoma and lung, breast, and head and neck cancers.446,448

Causal Evidence - Clinical Trial Data

Clarithromycin and gastric cancer: In contrast to its apparent mechanisms of action in other cancers, clarithromycin exerts its protective effects in gastric cancer through its antibacterial action against H. pylori. In a placebo-controlled trial that included 552 H. pylori-infected patients who were monitored for 10 years, one week of triple therapy with clarithromycin, amoxicillin (Amoxil), and omeprazole (Prilosec, a proton pump inhibitor) to eradicate H. pylori reduced gastric cancer risk.449 In a controlled trial that included 898 gastric cancer patients treated with surgery, those who received one week of clarithromycin, amoxicillin, and omeprazole to eradicate H. pylori were half as likely to develop new gastric cancer compared with those who did not receive triple therapy during a median follow-up period of almost six years.450 Another placebo-controlled trial in 396 surgically treated gastric cancer patients performed by the same research group found triple therapy with clarithromycin, amoxicillin, and rabeprazole (Pariet, another proton pump inhibitor) reduced the risk of subsequent new gastric cancer compared with placebo during a median of 5.9 years of monitoring.451 Unfortunately, because of widespread resistance to clarithromycin, its usefulness in reducing gastric cancer risk through H. pylori eradication may be limited.452

Clarithromycin and multiple myeloma: Several clinical trials using clarithromycin alone in patients with multiple myeloma have reported no survival benefits446,453; however, combinations of clarithromycin with the anti-inflammatory steroid dexamethasone (Decadron), with and without chemotherapy agents, have shown promising results in preliminary uncontrolled trials.446

Treatments for multiple myeloma that combine clarithromycin and dexamethasone with either lenalidomide (Revlimid) or thalidomide (Thalomid) appear to result in similar outcomes and may be more beneficial than protocols that do not include clarithromycin.454 One study compared outcomes from 72 patients who received clarithromycin plus lenalidomide and dexamethasone to treat newly diagnosed multiple myeloma to 72 similar patients who received lenalidomide and dexamethasone without clarithromycin. Among those receiving clarithromycin, 73.6% had a very good partial response or better, with 45.8% having a complete response; among those not receiving clarithromycin, only 33.3% had a very good partial response or better, and 13.9% had a complete response.455 A follow-up report after a median of 6.6 years found 68% of patients treated with clarithromycin continued to exhibit a very good partial response or better.456

An uncontrolled trial analyzing 50 patients with untreated or previously treated multiple myeloma or Waldenström’s macroglobulinemia found the combination of clarithromycin plus thalidomide and dexamethasone resulted in a 93% response rate, with 53% achieving a complete or near complete response.457 In 26 patients with newly diagnosed multiple myeloma, clarithromycin in combination with dexamethasone and both thalidomide and lenalidomide was associated with an overall response rate of 80% and four-year overall survival rate of 74.9%. In addition, 100% of 11 patients who also underwent a treatment known as autologous stem cell transplantation responded to treatment with clarithromycin plus chemotherapy.458

One study included 31 multiple myeloma patients and 17 patients with a related condition known as primary systemic light chain amyloidosis, all of whom had not responded to treatment with dexamethasone plus either thalidomide, lenalidomide, or pomalidomide (Pomalyst). The addition of clarithromycin to their treatment resulted in a response rate of 48% in multiple myeloma patients and 94% in amyloidosis patients.459 In an uncontrolled trial, 120 patients with relapsed or treatment-resistant multiple myeloma were given clarithromycin plus a standard chemotherapy protocol that included pomalidomide and dexamethasone. This combination resulted in an overall response rate of 60%, and 23% of patients achieved at least a very good partial response.460

In contrast, clarithromycin may increase toxicity of some chemotherapy agents. In a placebo-controlled trial in 58 patients with newly diagnosed multiple myeloma, the addition of clarithromycin to standard care with a chemotherapy protocol using cyclophosphamide, bortezomib, and dexamethasone resulted in reduced quality of life and severe adverse effects, leading to early termination of the trial.461,462

Clarithromycin and Waldenström’s macroglobulinemia: Waldenström’s macroglobulinemia is a rare type of lymphoplasmacytic lymphoma. Several reports suggest clarithromycin may enhance the efficacy of standard treatment for Waldenström’s macroglobulinemia. In a case report, an 84-year-old woman newly diagnosed with Waldenström’s macroglobulinemia responded to treatment with clarithromycin and the anti-inflammatory steroid prednisolone.463 A trial in 12 patients with Waldenström’s macroglobulinemia that had been unable to tolerate high-dose thalidomide therapy found the addition of clarithromycin and dexamethasone to low-dose thalidomide led to a partial treatment response in three patients (25%).464 In an uncontrolled trial, treatment with a combination of clarithromycin, dexamethasone, and thalidomide resulted in a significant response in 10 of 12 patients with treatment-resistant Waldenström’s macroglobulinemia after six weeks; however, all of the participants experienced neurological toxicity of varying intensities, which was severe enough to limit treatment in some cases.465

Clarithromycin and lymphoma: H. pylori has been implicated as a risk factor for mucosa-associated lymphoid tissue (MALT) lymphoma, a type of B-cell lymphoma that usually arises in the stomach.466 H. pylori eradication using triple therapy including clarithromycin has been found to induce long-term remission in a large majority of low- and high-grade MALT lymphoma patients.467-470

Clarithromycin may have benefits in MALT lymphoma that extend beyond its antibacterial effects. In an uncontrolled clinical trial, 23 H. pylori-negative patients with MALT lymphoma that recurred despite at least two previous treatments were given four 2-week courses of high-dose clarithromycin, with one week between each course. Complete remission occurred in six participants, partial response occurred in six others, and stabilization occurred in five. At a follow-up between 16 and 33 months after treatment, all participants survived and 13 (56%) had no sign of progression.471 In addition, a number of case reports indicate complete remission occurs in some patients with H. pylori -negative MALT lymphoma after treatment with clarithromycin.446

In a controlled trial in 55 patients with untreated, advanced stage, slow-growing, non-Hodgkin lymphoma, clarithromycin plus standard treatment with cyclophosphamide, vincristine (Oncovin), and prednisolone resulted in higher complete and overall response rates than standard treatment alone. The inclusion of clarithromycin also reduced levels of vascular endothelial growth factor, an important promoter of tumor-related blood vessel formation.472

Clarithromycin and lung cancer: In a controlled trial in 49 lung cancer patients who had already received chemotherapy, radiation therapy, or both, long-term treatment with clarithromycin increased survival time among those with non-small cell lung cancer.473 Another controlled trial in conventionally treated non-small cell lung cancer patients found the 33 participants given clarithromycin after chemotherapy and/or radiation therapy had reduced levels of the inflammatory marker interleukin-6 (IL-6), and this reduction was associated with increased survival time.474 Several other publications have reported on the effects of clarithromycin use in non-small cell lung cancer treatment.475-481 However, it is important to note that some evidence suggests clarithromycin may increase bone marrow suppression induced by the chemotherapy drugs docetaxel (Taxotere) and vinorelbine (Navelbine), causing dangerously decreased immune cell numbers, in lung cancer patients.482,483

Safety Considerations

Clarithromycin use is associated with mostly minor adverse side effects, particularly digestive symptoms, and in rare cases, it has been associated with more serious neurological symptoms. Because clarithromycin alters heart rhythm, it is not safe in patients with cardiac arrhythmias. There is also some evidence long-term clarithromycin use may increase cardiovascular risk. Clarithromycin may have negative interactions with some chemotherapy agents; therefore, care must be taken before combining clarithromycin with other cancer treatments.446

14 Doxycycline

Background

Doxycycline is a broad-spectrum, semisynthetic antibiotic from the tetracycline family that works by targeting ribosomal mechanisms for protein synthesis inside the cell.664-667 It is widely used to treat a host of bacterial infections, and is combined with antiparasitics for certain parasitic infections.668 Doxycycline has been proposed for use in cancer due to its antiproliferative effects in multiple cancer cell lines.664-666 It has also been found to inhibit tumor growth and proliferation in animal models of oral squamous cell carcinoma,669 hepatocellular carcinoma, breast cancer,670 and others.

A specific modification of mitochondrial transfer RNA (tRNA) (5-methylcytosine) has been identified as an essential requirement for metastasis of cancer cells. Without this modification, cancer cells are severely limited in their ability to spread. Of interest, inhibition of mitochondrial messenger RNA (mRNA) translation to tRNA with doxycycline treatment reduced the invasive metastatic spread of cancer cells in mice, suggesting that doxycycline’s metastasis-inhibiting properties could be due to inhibition of mRNA translation into 5-methylcytosine-containing tRNA.671

Causal Evidence – Clinical Trial Data

Doxycycline has not been extensively studied for improving cancer outcomes or survival, although several clinical trials are ongoing, using doxycycline alone or in combination with other drugs, including in breast, uterine,672,673 and pancreatic cancer.674

Cancer stem cells, which conventional cancer treatments like chemotherapy and radiation do not effectively eliminate, have been implicated in tumor recurrence, metastasis, and treatment failure in most cancer types.675 In one pilot study, 200 mg of doxycycline was administered for 14 days before surgery in nine women with breast cancer, while six served as untreated controls. Tumors from the doxycycline-treated group had significantly decreased “stemness” markers, indicating the targeting of cancer stem cells. However, doxycycline did not appear to affect markers of proliferation.676 One of the ongoing clinical trials is testing whether doxycycline can target cancer stem cells in people with pancreatic cancer.674

Doxycycline has mainly been studied as a treatment for epidermal growth factor receptor (EGFR) inhibitor-induced skin toxicity. EGFR inhibitors are essential treatments for a variety of cancers, including non-small cell lung cancer, colorectal cancer, pancreatic cancer, and others. However, EGFR inhibitors cause adverse cutaneous reactions, which often makes treatment highly uncomfortable and distressing for patients.677 Doxycycline and other tetracyclines have been shown in several—though not all—trials to be effective prophylaxis for this skin toxicity.677-680 Doxycycline treatment (100 mg) did not affect overall survival time in patients with locally advanced or metastatic non-small cell lung cancer680 or metastatic colorectal cancer,678 although survival was not a primary outcome in these studies.

Doxycycline has also been studied as a treatment for malignant pleural effusion (buildup of fluid and cancer cells between the chest wall and the lung), although again results have not shown consistent benefit.681,682 Further studies are necessary to elucidate doxycycline’s role, if any, in cancer therapy.

Safety Considerations

Doxycycline is generally well tolerated, with mild gastrointestinal side effects at typical doses. Digestive, photosensitivity, or skin reactions (sometimes severe) may occur. Systemic toxicity from doxycycline is rare.683

15 Itraconazole

Background

Itraconazole (Sporanox) is classified as a triazole antifungal medication and is widely used to treat fungal infections, including serious lung infections such as aspergillosis, blastomycosis, and histoplasmosis, as well as fungal infections of the fingernails and toenails (onychomycosis). It is also used to prevent fungal infections in immunocompromised patients such as those undergoing cancer therapies.484

Itraconazole has been shown to suppress cancer growth by inhibiting a gene signaling pathway called the hedgehog pathway,485 which transmits signals between the cell membrane and nucleus, and affects activation and repression of genes, including oncogenes.486 The hedgehog pathway serves important functions during development but is mostly inactive in adult cells; abnormal activation of the hedgehog pathway, possibly due to epigenetic alteration, has been implicated in the development of various types of cancer, such as lung, skin, endometrial, cervical, brain, breast, gastric, pancreatic, ovarian, and liver cancers.485,486 Evidence suggests itraconazole also inhibits other key signaling pathways, including the mTOR pathway.484,487 By suppressing these cell-signaling pathways, itraconazole arrests cancer cell proliferation, stimulates cancer cell apoptosis, and inhibits tumor-initiated angiogenesis (formation of new blood vessels), and may increase cancer cell sensitivity to chemotherapy agents.484,485

In laboratory and animal studies, itraconazole has demonstrated anti-cancer effects against brain,488 breast,489,490 cervical,491 endometrial,492,493 ovarian,494 oral,495 esophageal,496 pancreatic,497 gastric,498 and colorectal39,499 cancers, as well as melanoma.500 In an animal model of Barrett’s esophagus, treatment with itraconazole lowered the occurrence of esophageal cancer from 32% to 8%.501 In gastric cancer cells, itraconazole increased susceptibility to chemotherapy with 5-FU.502 A combination of itraconazole plus doxorubicin (Adriamycin) was more effective than either drug alone in a study in acute myeloid leukemia cells.503

Observational (Correlational) Evidence

In a study that included data from 60 gastric cancer patients who underwent treatment that included 5-FU, those who incidentally received itraconazole (200–400 mg per day intravenously for four to five days) had a higher treatment response rate (59%) compared with those who did not (45%). They also had longer progression-free and overall survival.502 Another study analyzed survival data collected during a placebo-controlled trial to evaluate the effect of prophylactic itraconazole on fungal infections in patients being treated with chemotherapy for acute lymphoblastic leukemia (ALL) and acute myelogenous leukemia (AML). The study found small improvements in outcomes among those treated with itraconazole compared with those treated with placebo, suggesting itraconazole may have increased chemotherapy sensitivity.504

One research group examined outcomes in cancer patients who had relapsed or persistent cancer despite initial treatment with chemotherapy, and whose second-line treatment included chemotherapy plus itraconazole to prevent fungal infections:

  • In 38 relapsed pancreatic cancer patients, a response rate of 37% was observed: one subject experienced a complete response and 13 experienced partial responses.505
  • In 28 patients with relapsed metastatic biliary tract cancer, a response rate of 57% was reported: two subjects had complete responses and 14 had partial responses.506
  • In 13 breast cancer patients whose conditions had worsened during chemotherapy, the response rate was 62%.507
  • In nine patients with recurrent or persistent ovarian cancer, the response rate was 44%.508
  • Among 55 patients with ovarian cancer that was unresponsive to chemotherapy, those who received itraconazole had longer overall and progression-free survival.509

Although these were retrospective observations, in most cases lacking controls for comparison, they indicate itraconazole has possible benefits in patients with advanced and progressive cancers,510 and provide data that can be used in the future to evaluate whether the addition of itraconazole to second-line chemotherapy improves treatment responsiveness and lengthens survival.

Causal Evidence - Clinical Trial Data

The following describes the body of clinical evidence as of mid-2020 regarding itraconazole use in cancer therapy. More clinical trials investigating the potential anti-cancer benefits of itraconazole, mainly as an adjuvant to standard cancer therapies, are underway as of the time of this writing.487

Cumulatively, this evidence suggests itraconazole has the potential to favorably affect cancer treatment outcomes beyond its ability to prevent treatment-related fungal infections. Nevertheless, more rigorous controlled trials in patients with a range of cancers at various stages of severity are needed to confirm and clarify its usefulness.

Itraconazole and basal cell carcinoma: A randomized controlled trial included 29 patients with basal cell carcinoma (a type of skin cancer), each with one or more tumors greater than 4 mm in diameter. Fifteen of the subjects received 200 mg itraconazole twice daily for one month; four received 100 mg itraconazole twice daily for an average of 2.3 months; and 10 received no itraconazole. In those treated with itraconazole, cancer cell proliferation was reduced by 45% and tumor area by 24%, whereas neither of these parameters changed in the untreated group. Among the eight subjects with multiple tumors, four had a partial response and four had disease stabilization.511 In a study that reported on five cases of metastatic basal cell carcinoma, treatment with itraconazole and arsenic trioxide (another hedgehog-inhibiting agent) led to stabilization of cancer in three subjects.512 Another report described the case of an 87-year-old man with recurrent metastatic basal cell carcinoma with multiple tumors in the head. After exhausting other treatment options, the patient was given itraconazole plus sonidegib (Odomzo), another hedgehog inhibitor with a slightly different mechanism of action. After eight months on this treatment regime, one tumor resolved completely and the others stabilized.513

Itraconazole and prostate cancer: In an open-label trial, 46 men with metastatic prostate cancer were treated with either low-dose (200 mg per day) or high-dose (600 mg per day) itraconazole. Forty-eight percent of the high-dose and 11.8% of the low-dose patients achieved stable PSA levels (rising less than 25% from baseline) at week 24; in addition, high-dose treatment resulted in longer progression-free survival compared with low-dose treatment.514 Another trial enrolled 21 participants in whom lab tests indicated prostate cancer had become active again after treatment. The participants had PSA doubling times (the time it takes for the PSA level to double) ranging from 1.2 to 13 months. Nineteen of the subjects completed 12 weeks of treatment with itraconazole: PSA doubling time decreased by an average of 25% in nine subjects and did not change significantly in the remaining 10 subjects. Additionally, whereas other treatment options are known to cause adverse side effects related to reduced testosterone levels, itraconazole had no effect on testosterone levels.515 In a case report describing a 65-year-old man with lab tests indicating recurrence of prostate cancer, 600 mg itraconazole per day led to progressive reductions in PSA levels for five months, at which point adverse side effects required him to discontinue treatment.516

Itraconazole and stomach cancer: In a pilot trial, 23 patients with advanced or recurrent gastric cancer that was not surgically treatable received a treatment regime with nab-paclitaxel (Abraxane), oxaliplatin (Eloxatin), and itraconazole, administered in six to eight cycles; two patients had a complete response and 14 had partial responses.517

Itraconazole and pancreatic cancer: A case was reported of a 64-year-old man with advanced pancreatic cancer that was not amenable to surgery. Treatment with chemotherapy and radiation therapy was not successful; however, nine months of itraconazole use for a fungal infection (a result of treatment-related immune suppression) corresponded with tumor regression such that surgery was feasible.518

Itraconazole and lung cancer: In a controlled trial in 23 patients with non-small cell lung cancer that was progressing despite previous treatment, those treated with the chemotherapy agent pemetrexed (Alimta) plus itraconazole were more likely to have no progression after three months and had longer progression-free and overall survival times than those treated with pemetrexed alone.519 In another clinical trial, 13 patients with non-small cell lung cancer received 600 mg per day of itraconazole for 10 to 14 days prior to surgery to remove their tumors. This treatment led to reduced tumor volume and tumor blood flow at the time of surgery, and the effects were stronger in those whose tumors had higher concentrations of itraconazole.520

Safety Considerations

Itraconazole has been safely used for decades and is generally well tolerated, but adverse effects can occur, particularly with long-term use. In a minority of short-term itraconazole users, side effects such as nausea and other digestive problems, rashes, fatigue, dizziness, and neuropathy can occur,484 whereas protracted itraconazole use may cause liver toxicity, heart failure, low white blood cell count, hair loss, decreased libido, breast tissue enlargement in men, impotence, low sperm count, low levels of potassium or sodium, pancreatitis, and rarely adrenal insufficiency.484,521 In addition, because itraconazole interacts with a major metabolic pathway in the liver, it can cause numerous drug interactions.522

16 Mebendazole

Background

Mebendazole (Vermox), a medication used to treat infections caused by helminths (parasitic worms), has been under investigation for possible anti-cancer effects since it was discovered to inhibit tubulin polymerization, an action it shares with some conventional chemotherapy drugs.523 Tubulin inhibitors interfere with the process of linking tubulin proteins (polymerization) to form important cellular structures called microtubules. Microtubules are components of the intracellular matrix and are needed for maintaining cell shape and function, having roles in cell signaling, movement of molecules within the cell, formation of blood vessels, and cell division. By affecting microtubule formation, tubulin inhibitors can slow or stop cancer cell proliferation and promote apoptosis.524

Laboratory studies have shown mebendazole reduces cancer cell proliferation, invasion, and migration; inhibits critical cancer cell signaling pathways; interrupts tumor-induced formation of new blood vessels; and supports anti-cancer immune cell function.523,525-527 Through these actions, mebendazole appears to block tumor growth and spread, induce apoptosis, and increase sensitivity to other anti-cancer therapies.523,528 In particular, mebendazole has demonstrated promising anti-cancer effects in laboratory and animal models of brain cancer.529-532 A study in tumor cells from brain cancer patients found certain patterns of gene expression were associated with poor response to the standard chemotherapy drug temozolomide, and the addition of mebendazole and another chemotherapy drug enhanced susceptibility.533 Another study in cancer cells from women with triple-negative breast cancer found mebendazole increased their vulnerability to radiation therapy.534 Mebendazole has also been found to have positive effects in preclinical models of a broad range of cancers, including gastric,535,536 colorectal,537,538 prostate,539 breast,540 liver,541 lung,542 thyroid,543 head and neck,544 gallbladder,545 and adrenal cortex546 cancers, as well as ALL,547-549 melanoma,550,551 myeloma,552 and meningioma553; however, one study found it was not effective in a rodent model of fibrosarcoma.554

Observational (Correlational) Evidence

While there are no completed clinical trials investigating the potential role of mebendazole in cancer treatment, such trials are underway as of mid-2020.523,555 Two case reports, however, provide early support for its potential benefits.556,557

Mebendazole and cancer of the adrenal gland: A case was reported of a 48-year-old man with metastatic cancer of the adrenal cortex, a rare cancer with few treatment options. After finding the cancer was progressing despite treatment attempts with multiple chemotherapy agents, the patient was given 100 mg mebendazole twice daily with no other therapy. He remained stable, with no cancer progression, for two years.556

Mebendazole and colorectal cancer: A case was reported in which a 74-year-old patient with metastatic colon cancer was experiencing disease progression despite multiple treatment attempts using chemotherapy protocols. Six weeks after starting 200 mg mebendazole per day as a stand-alone therapy, the patient had complete remission of metastatic tumors in the lungs and lymph nodes and had partial remission in the liver. Treatment was temporarily halted due to lab test abnormalities indicating liver toxicity and then resumed at half dose. The lab tests slowly normalized and the patient and disease remained stable.557

Mebendazole and brain cancer: In a preliminary dose-finding trial, patients with relapsed brain cancer were treated with chemotherapy and mebendazole, with or without radiation therapy. This study found mebendazole was tolerable at very high doses, but could not provide useful information about its effects on outcomes.558

Safety Considerations

Mebendazole is a well-tolerated drug with mainly gastrointestinal side effects at typical doses. It has also been reported to trigger allergic reactions in rare cases.555

17 All-Trans Retinoic Acid

Background

All-trans retinoic acid (ATRA) is a member of the retinoid family and the most active metabolite of vitamin A.559 Dietary vitamin A, after a series of metabolic changes in the gut and bloodstream, is converted to ATRA inside the body’s cells, where it can be transported into the nucleus and play a role in regulating gene transcription. The rate of its production and breakdown is controlled within the cells.559-561 The drug version of ATRA, tretinoin (Vesanoid, Retin-A), is best known for its topical and oral use in treating skin conditions such as acne, psoriasis, ichthyosis (a genetic skin disorder characterized by dry, thickened, scaly skin), and skin cancer.562 ATRA has demonstrated a number of anti-cancer actions, and research interest in its potential role in cancer therapy has grown over the past decade.559,561

Humans require 700–900 mcg vitamin A (or retinol equivalents of pro-vitamin A carotenoids such as beta-carotene) from the diet each day in order to support normal cellular function, but therapeutic doses of ATRA are many-fold higher.559 Most dietary vitamin A is stored in the liver, from where it is slowly released into circulation.563 Because vitamin A is non-water-soluble, its transport through the blood and uptake by cells are facilitated and regulated by retinol binding proteins and cell membrane receptors; once inside the cell, its conversion to ATRA is enzyme-dependent.559,561 The capacities of retinol binding proteins, receptors, and enzymes limit the extent to which increasing vitamin A intake can raise intracellular ATRA concentrations. Furthermore, exceeding these capacities can result in toxic damage to cell membranes and disrupt oxidation-reduction balance.564 ATRA activity and breakdown is also controlled by specific binding proteins and nuclear membrane receptors.

ATRA is an important inducer of normal cell differentiation, as well as apoptosis.560 In vitro experiments have associated deficient levels of intracellular ATRA with cancer initiation and progression, and ATRA exhibits numerous mechanisms that could suppress cancer proliferation and metastasis.559,562,565 In fact, tumor cells frequently have altered retinoid metabolism and diminished ATRA signaling, and treatment with ATRA has inhibited cancer growth in a number of in vitro models.559,563 In addition to promoting cell differentiation and apoptosis in tumor cells, ATRA appears to normalize the function of cancer stem cells.559,566 It is also an important stabilizer of tight junctions (interconnections between neighboring cells), which may reduce the ability of cancer cells to metastasize.559 Preclinical research indicates ATRA has potential synergistic effects with other anti-cancer agents.567

ATRA is used in combination with other chemotherapeutics as the standard of care for acute promyelocytic leukemia (a blood cancer) and neuroblastoma (a nerve cell cancer that affects children).559,568 In laboratory research, ATRA has also been shown to have anti-cancer activity against thyroid,569-572 breast,573 cervical,574,575 ovarian,576,577 liver,578 colon,579-581 gastric,582 lung,583,584 pancreatic,585-587 brain,588 and esophageal589,590 cancers, as well as melanoma,591 non-melanoma skin cancer,592-594 acute myeloid leukemia,595 lymphoma,596-598 and chordoma (a type of bone cancer).599,600

Although ATRA’s performance in clinical trials to date has been mostly disappointing,560 more than 20 clinical trials in patients with various types of cancer are actively under way as researchers continue to investigate its best use.601 A few key challenges to its utility as an anti-cancer agent have been identified: ATRA is a fatty compound with low water solubility, limiting its absorption via the gut; it is rapidly metabolized and inactivated once absorbed; and cancer cells frequently have or develop retinoid resistance.559,560 Strategies to overcome these challenges are incorporated in a number of new ATRA formulations and delivery methods currently being developed and investigated.559 Except where otherwise specified, the clinical evidence described below is based on the use of oral (systemic) ATRA.

Causal Evidence – Clinical Trial Data

ATRA and acute promyelocytic leukemia: Acute promyelocytic leukemia is a severe leukemia which, before the introduction of ATRA, carried a very poor prognosis but is now highly curable. It is characterized by proliferation of immature white blood cell precursors called promyelocytes and rapid deterioration of immune function.602 ATRA, alone or in combination with arsenic trioxide, repairs an epigenetic alteration associated with acute promyelocytic leukemia and forces either differentiation or apoptosis of leukemic promyelocytes. ATRA can stimulate complete remission in a large majority of acute promyelocytic leukemia patients, and has been a mainstay of conventional acute promyelocytic leukemia treatment since its approval by the FDA in 1995.560,603,604

ATRA and neuroblastoma: Neuroblastoma is a group of pediatric cancers that arise from nervous system tissue. ATRA, or another retinoid, cis-retinoic acid, is a standard part of the maintenance phase of chemotherapy for neuroblastoma.559,605 For this purpose, the retinoid is generally combined with the monoclonal antibody dinutuximab (Unituxin).606

ATRA and myelodysplastic syndromes and acute myeloid leukemia: Several mostly older clinical trials have examined the effect of ATRA in patients with myelodysplastic syndromes and acute myeloid leukemia. In a randomized controlled trial in 242 patients with acute myeloid leukemia aged 61 years and older, those given ATRA in conjunction with standard chemotherapy had significantly better treatment response rates and overall survival than those given standard care alone.607 In a controlled trial, 45 poor prognosis patients in remission after treatment for myelodysplastic syndromes or acute myeloid leukemia were placed on a maintenance program with an alternating medication schedule, one arm of which included ATRA and the other another retinoid. After a median of 52 months of monitoring, fewer relapses and better disease-free survival were noted in the treated group versus 49 similar patients who received no maintenance therapy.608 In an open trial in 53 patients affected by myelodysplastic syndromes or acute myeloid leukemia with poor prognosis, a combination of 5-azacitidine, valproic acid, and ATRA was found to be safe and appeared to have clinical benefits.609 In another trial, myelodysplastic syndromes or acute myeloid leukemia patients who responded to initial treatment with valproic acid but later relapsed had an improved second response after the addition of ATRA to treatment, but the addition of ATRA to treatment did not improve outcomes in other myelodysplastic syndromes or acute myeloid leukemia patients.610 A number of other trials have found no benefit from adding ATRA to myelodysplastic syndromes and acute myeloid leukemia treatments.337,611-614 A meta-analysis of eight trials that included a combined total of almost 4,000 patients found the addition of ATRA to treatment of acute myeloid leukemia did not alter treatment response or survival outcomes.615

ATRA and various solid tumors: After ATRA was found to be effective in treatment of acute promyelocytic leukemia, several research teams conducted phase I dose-finding trials in cancer patients with various advanced solid tumors. Unfortunately, no major tumor responses were observed in these trials, and many side effects occurred.560,561 In addition, in an early pilot trial in pediatric cancer patients, no positive effects were seen in children with solid tumors, and several instances of increased intercranial pressure, which resolved when ATRA therapy was discontinued, were reported.616

ATRA and specific solid tumors: Despite the lack of clear benefit in small phase I trials in patients with various types of solid tumors, other trials have gone on to investigate the potential usefulness of ATRA in groups of patients with specific types of solid tumors.

ATRA and pancreatic cancer: A phase I clinical trial that included 27 patients with advanced or metastatic pancreatic cancer who received no prior treatment for their cancer published its results in 2020. Patients received ATRA in combination with the chemotherapy agents gemcitabine and nab-paclitaxel. The results of the trial indicated the addition of ATRA to standard chemotherapy did not increase toxicity. Although it was not a controlled trial designed to evaluate outcomes, the results of this trial were considered promising.617

ATRA and thyroid cancer: One older study reported on the results of ATRA therapy in 11 patients with thyroid cancers. All the subjects had advanced disease that had been unsuccessfully treated with surgery and radioiodine therapy. Lack of sufficient uptake of radioiodine was one reason preventing further standard treatment. After treatment with ATRA, four subjects had increased radioiodine uptake, five had a partial treatment response, and two had stabilization of their condition.618

ATRA and cervical cancer: Topical ATRA has been a treatment option for decades in women with cervical intraepithelial neoplasia (CIN, a precancerous condition), and was reported in an older trial to increase remission rates in those with stage II and milder CIN.619 In another older clinical trial in 26 women with metastatic or recurrent cervical cancer, the combination of interferon-alpha (IFN-α) (Intron A) plus oral ATRA, whether on daily or intermittent schedules, resulted in no response. Because blood levels of ATRA were sufficiently elevated in those receiving intermittent treatment, the study authors speculated the lack of response was likely due to cancer cell resistance to the anti-cancer effects of ATRA.620 A more recent placebo-controlled trial in 175 women with CIN II and III found topical ATRA had no effect on outcomes.620

Two other clinical trials examined the effect of ATRA in combination with IFN-α and the chemotherapy agent cisplatin. In both trials, toxicity limited therapy in the majority of participants. However, researchers in these trials concluded that the response rate was reason to consider further trials of retinoids in combination therapy in early disease.621,622

ATRA and breast cancer: In an open trial, 17 women with previously treated metastatic or recurrent breast cancer were treated with ATRA before and during chemotherapy with paclitaxel. The outcomes were reported to be similar to those expected with paclitaxel alone.623 In an early open trial, ATRA alone was also found to have no clinical effect in 17 women with metastatic breast cancer that was unresponsive to hormone therapy.624

ATRA may have a role as an adjunct to tamoxifen (Soltamox) in women who have had breast cancer. In another early open trial that included 25 patients with advanced, potentially hormone-sensitive breast cancer who were treated with tamoxifen plus ATRA, some women experienced promising clinical responses, including some who had progressed when treated with tamoxifen alone.625 In a randomized controlled trial in 30 women with locally advanced breast cancer, participants were treated with ATRA, ATRA plus tamoxifen, or tamoxifen alone for 21 days prior to surgery for tumor removal. While tamoxifen use caused increased blood levels of pro-clotting markers, ATRA did not cause this effect and suppressed the rise in clotting markers when used in combination with tamoxifen. An increase in clot risk is an important side effect of tamoxifen, and this research suggests ATRA may mitigate this problem.626

ATRA and lung cancer: Several clinical trials have evaluated the effect of ATRA in advanced or metastatic non-small cell lung cancer. Two early trials found ATRA alone had low toxicity but was not associated with significant improvement in outcomes.627,628 In two early open trials, combination treatment with ATRA plus cisplatin (20 participants) and ATRA plus IFN-α (29 participants) was reported to have resulted in modest clinical responses.629,630 In a placebo-controlled trial in 107 participants being treated with paclitaxel and cisplatin for advanced non-small cell lung cancer, adding ATRA to treatment increased response rate and prolonged progression-free survival.631 In an early trial in patients with advanced small cell lung cancer, the addition of ATRA to chemotherapy with cisplatin and etoposide (Vepesid, Toposar) was poorly tolerated, resulting in early discontinuation of ATRA in most patients.632

One placebo-controlled trial evaluated the potential for ATRA to reduce nerve problems related to chemotherapy in lung cancer patients: the trial included 95 patients being treated with cisplatin and paclitaxel for non-small cell lung cancer, and found ATRA reduced chemotherapy-induced nerve damage, as well as the risk of moderate-to-severe neuropathy.633

ATRA and melanoma: A controlled trial with 10 participants showed ATRA decreased levels of myeloid-derived suppressor cells in advanced melanoma patients being treated with the monoclonal antibody ipilimumab (Yervoy). Myeloid-derived suppressor cells are tumor-induced immune cells that contribute to resistance to immunotherapies; therefore, these findings suggest ATRA may have a role as an adjunct to immunotherapy in melanoma treatment.634 An early open trial found the combination of ATRA plus IFN-α was not more effective than IFN-α alone in 52 patients with metastatic melanoma.635

Several trials have looked at the effect of topical ATRA on dysplastic nevi, which are precursors to melanoma. One research group reported partial or complete resolution of some dysplastic nevi (irregular mole-like skin lesions) with topical ATRA. However, this was an uncontrolled setting and thus the evidence may not be reliable.636 In an early controlled trial, five men with a total of 32 dysplastic nevi received topical ATRA on half of their lesions and the remaining lesions were untreated for six months. At the end of the trial, four of 16 treated nevi and 13 of 16 untreated nevi remained dysplastic, but the use of ATRA also caused significant skin irritation.637 In another early controlled trial with 30 participants with dysplastic nevi, ATRA alone and when combined with topical hydrocortisone (which reduced treatment-related skin irritation) resulted in mild improvement in dysplastic cell characteristics, but not enough to preclude the need for surgical removal.638

ATRA and non-melanoma skin cancers: ATRA and other retinoids have long been used to prevent and treat various types of non-melanoma skin cancers.639 One case report described an 86-year-old Caucasian man with an invasive squamous cell carcinoma for whom treatment consisted of a topical solution containing ATRA, imiquimod (Aldera), and 5-FU (Efudex) on five days per week for a total of 24 treatments, along with intermittent topical cryotherapy (the use of extreme cold to destroy diseased tissue). Fifteen months after the end of treatment, there was no sign of residual cancer.640

A research review found the majority of studies indicate retinoid therapy, including topical ATRA, can reduce the number of precancerous actinic keratoses.641 A placebo-controlled trial examined the potential for topical ATRA to reduce the risk of the two most common skin cancers, squamous cell carcinoma and basal cell carcinoma, in 1,131 individuals with a history of two or more cancerous skin lesions on the face or ears in the previous five years. No difference in risk of new skin cancers was seen after 1.5–5.5 years of monitoring.642 Interestingly, the tretinoin group experienced a higher death rate than the placebo group; although the relationship was deemed unlikely to be causal, there was no clear explanation for the difference and the trial was halted six months early.643

ATRA and liver cancer: An early open clinical trial that included 15 patients with inoperable hepatocellular carcinoma reported treatment with ATRA in combination with vitamin E and tamoxifen appeared to prolong survival.644 However, another early open trial in 29 hepatocellular carcinoma patients found ATRA alone had no benefit and high-grade toxic side effects were noted in 15 participants.645

ATRA and gastric cancer: In a randomized controlled trial in 122 patients with gastric dysplasia (a precancerous condition), those given ATRA plus standard treatment with a proton pump inhibitor (omeprazole) and a protectant (sucralfate [Carafate]) experienced greater reduction in dysplasia compared with those given standard treatment alone.646

ATRA and kidney cancer: An early phase I clinical trial showed the combination of ATRA plus IFN-α was reasonably safe in patients with advanced renal cell carcinoma, and outcomes suggested some possible clinical benefit.647

Safety Considerations

ATRA is a drug with a serious side effect profile, and thus carries a strong label warning that it should be used in acute promyelocytic leukemia only under the close supervision of health care practitioners experienced in managing acute leukemia in the context of an appropriate facility or clinical setting. Its best recognized possible major severe adverse effects are a dangerous syndrome marked by fever, shortness of breath, and impaired function or failure of multiple organ systems, called acute promyelocytic leukemia differentiation syndrome (APL DS), which occurs in about 25% of patients with acute promyelocytic leukemia treated with ATRA and which can be serious or fatal; severe high white blood cell count, called leukocytosis, which occurs in roughly 40% of ATRA-treated patients, which also can be serious or fatal; and teratogenicity (causing birth defects) at all stages of pregnancy, especially in the first trimester. Individuals using ATRA are warned to initiate treatment within one week of a negative pregnancy test and use two safe contraceptive methods until one month after ATRA is discontinued.568

Many chemotherapy drugs target rapidly-dividing cells and frequently cause toxicity by damaging rapidly-dividing non-cancerous cells, such as immune cells and cells that line the intestines; conversely, ATRA reactivates signaling pathways that are suppressed during the development of cancer, forcing cancer cells to resume normal cell cycles or die through apoptosis without damaging non-cancerous cells.648

Other common adverse side effects associated with oral ATRA use include increased triglyceride and liver enzyme levels, skin problems, dryness of mucous membranes, nausea, vomiting, and headache. At higher doses and mainly in children, increased intracranial pressure (a condition known as pseudotumor cerebri) has been reported.560 Topical ATRA use has been associated with skin burning, itching, and irritation.649

2022

  • Aug: Updated section on metformin and liver cancer in Metformin
  • Jul: Added section on doxycycline
  • Jun: Updated section on metformin and breast cancer in Metformin

2021

  • Jun: Updated section on statins and colorectal cancer in Statin Drugs
  • Feb: Initial publication

Disclaimer and Safety Information

This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.

The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.

  1. Antoszczak M, Markowska A, Markowska J, Huczynski A. Old wine in new bottles: Drug repurposing in oncology. European journal of pharmacology. Jan 5 2020;866:172784. doi:10.1016/j.ejphar.2019.172784
  2. Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP, Vikas P. The Repurposing Drugs in Oncology (ReDO) Project. Ecancermedicalscience. 2014;8:442. doi:10.3332/ecancer.2014.442
  3. Corsello SM, Nagari RT, Spangler RD, et al. Discovering the anticancer potential of non-oncology drugs by systematic viability profiling. Nature Cancer. 2020/02/01 2020;1(2):235-248. doi:10.1038/s43018-019-0018-6
  4. Nowak-Sliwinska P, Scapozza L, Ruiz I Altaba A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim Biophys Acta Rev Cancer. 2019;1871(2):434-454. doi:10.1016/j.bbcan.2019.04.005
  5. Metformin for Prediabetes. JAMA. 2017;317(11):1171-1171. doi:10.1001/jama.2016.17844
  6. Aljofan M, Riethmacher D. Anticancer activity of metformin: a systematic review of the literature. Future Sci OA. Aug 22 2019;5(8):Fso410. doi:10.2144/fsoa-2019-0053
  7. Vancura A, Bu P, Bhagwat M, Zeng J, Vancurova I. Metformin as an Anticancer Agent. Trends Pharmacol Sci. Oct 2018;39(10):867-878. doi:10.1016/j.tips.2018.07.006
  8. Wrobel MP, Marek B, Kajdaniuk D, Rokicka D, Szymborska-Kajanek A, Strojek K. Metformin - a new old drug. Endokrynologia Polska. 2017;68(4):482-496. doi:10.5603/ep.2017.0050
  9. Samuel SM, Varghese E, Kubatka P, Triggle CR, Busselberg D. Metformin: The Answer to Cancer in a Flower? Current Knowledge and Future Prospects of Metformin as an Anti-Cancer Agent in Breast Cancer. Biomolecules. Dec 9 2019;9(12)doi:10.3390/biom9120846
  10. Saraei P, Asadi I, Kakar MA, Moradi-Kor N. The beneficial effects of metformin on cancer prevention and therapy: a comprehensive review of recent advances. Cancer management and research. 2019;11:3295-3313. doi:10.2147/cmar.S200059
  11. Kurelac I, Umesh Ganesh N, Iorio M, Porcelli AM, Gasparre G. The multifaceted effects of metformin on tumor microenvironment. Seminars in cell & developmental biology. Feb 2020;98:90-97. doi:10.1016/j.semcdb.2019.05.010
  12. Li M, Li X, Zhang H, Lu Y. Molecular Mechanisms of Metformin for Diabetes and Cancer Treatment. Front Physiol. 2018;9:1039. doi:10.3389/fphys.2018.01039
  13. Andrzejewski S, Siegel PM, St-Pierre J. Metabolic Profiles Associated With Metformin Efficacy in Cancer. Frontiers in endocrinology. 2018;9:372. doi:10.3389/fendo.2018.00372
  14. Bahrambeigi S, Shafiei-Irannejad V. Immune-mediated anti-tumor effects of metformin; targeting metabolic reprogramming of T cells as a new possible mechanism for anti-cancer effects of metformin. Biochemical pharmacology. Apr 2020;174:113787. doi:10.1016/j.bcp.2019.113787
  15. Kunisada Y, Eikawa S, Tomonobu N, et al. Attenuation of CD4(+)CD25(+) Regulatory T Cells in the Tumor Microenvironment by Metformin, a Type 2 Diabetes Drug. EBioMedicine. Nov 2017;25:154-164. doi:10.1016/j.ebiom.2017.10.009
  16. Huang QY, Yao F, Zhou CR, et al. Role of gut microbiome in regulating the effectiveness of metformin in reducing colorectal cancer in type 2 diabetes. World journal of clinical cases. Dec 26 2020;8(24):6213-6228. doi:10.12998/wjcc.v8.i24.6213
  17. Ma W, Chen J, Meng Y, Yang J, Cui Q, Zhou Y. Metformin Alters Gut Microbiota of Healthy Mice: Implication for Its Potential Role in Gut Microbiota Homeostasis. Frontiers in microbiology. 2018;9:1336-1336. doi:10.3389/fmicb.2018.01336
  18. Jones GR, Molloy MP. Metformin, Microbiome and Protection Against Colorectal Cancer. Dig Dis Sci. Jun 12 2020;doi:10.1007/s10620-020-06390-4
  19. Prattichizzo F, Giuliani A, Mensà E, et al. Pleiotropic effects of metformin: Shaping the microbiome to manage type 2 diabetes and postpone ageing. Ageing Res Rev. Dec 2018;48:87-98. doi:10.1016/j.arr.2018.10.003
  20. Rodriguez J, Hiel S, Delzenne NM. Metformin: old friend, new ways of action-implication of the gut microbiome? Current opinion in clinical nutrition and metabolic care. Jul 2018;21(4):294-301. doi:10.1097/mco.0000000000000468
  21. Deng M, Lei S, Huang D, et al. Suppressive effects of metformin on colorectal adenoma incidence and malignant progression. Pathol Res Pract. Feb 2020;216(2):152775. doi:10.1016/j.prp.2019.152775
  22. Wang Y, Xiao J, Zhao Y, Du S, Du J. Effect of metformin on the mortality of colorectal cancer patients with T2DM: meta-analysis of sex differences. International journal of colorectal disease. Feb 25 2020;doi:10.1007/s00384-020-03539-5
  23. Powell MK, Cempirkova D, Dundr P, et al. Metformin Treatment for Diabetes Mellitus Correlates with Progression and Survival in Colorectal Carcinoma. Translational oncology. Feb 2020;13(2):383-392. doi:10.1016/j.tranon.2019.10.011
  24. Kim JM, Park JW, Lee JH, et al. Survival Benefit for Metformin Through Better Tumor Response By Neoadjuvant CCRT in Rectal Cancer. Diseases of the colon and rectum. Feb 26 2020;doi:10.1097/dcr.0000000000001624
  25. Roshan MH, Shing YK, Pace NP. Metformin as an adjuvant in breast cancer treatment. SAGE open medicine. 2019;7:2050312119865114. doi:10.1177/2050312119865114
  26. Lu MZ, Li DY, Wang XF. Effect of metformin use on the risk and prognosis of ovarian cancer: an updated systematic review and meta-analysis. Panminerva medica. Jul 8 2019;doi:10.23736/s0031-0808.19.03640-1
  27. Shi J, Liu B, Wang H, Zhang T, Yang L. Association of metformin use with ovarian cancer incidence and prognosis: a systematic review and meta-analysis. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society . Jan 2019;29(1):140-146. doi:10.1136/ijgc-2018-000060
  28. Wang Y, Liu X, Yan P, Bi Y, Liu Y, Zhang ZJ. No Effect of Metformin on Ovarian Cancer Survival: A Systematic Review and Meta-Analysis of Cohort Studies. Curr Pharm Des. 2019;25(23):2595-2601. doi:10.2174/1381612825666190716113126
  29. Cunha V, Cotrim HP, Rocha R, Carvalho K, Lins-Kusterer L. Metformin in the prevention of hepatocellular carcinoma in diabetic patients: A systematic review. Ann Hepatol. Nov 28 2019;doi:10.1016/j.aohep.2019.10.005
  30. Zhou J, Ke Y, Lei X, et al. Meta-analysis: The efficacy of metformin and other anti-hyperglycemic agents in prolonging the survival of hepatocellular carcinoma patients with type 2 diabetes. Ann Hepatol. Dec 16 2019;doi:10.1016/j.aohep.2019.11.008
  31. Zhang P, Li H, Tan X, Chen L, Wang S. Association of metformin use with cancer incidence and mortality: a meta-analysis. Cancer epidemiology. Jun 2013;37(3):207-18. doi:10.1016/j.canep.2012.12.009
  32. Li J, Liu R, Sun Z, et al. The association between statin use and endometrial cancer survival outcome: A meta-analysis. Medicine. Nov 2018;97(47):e13264. doi:10.1097/md.0000000000013264
  33. Wan G, Sun X, Li F, et al. Survival Benefit of Metformin Adjuvant Treatment For Pancreatic Cancer Patients: a Systematic Review and Meta-Analysis. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology . 2018;49(3):837-847. doi:10.1159/000493214
  34. Toriola AT, Luo S, Thomas TS, et al. Metformin Use and Pancreatic Cancer Survival among Non-Hispanic White and African American U.S. Veterans with Diabetes Mellitus. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Jan 2020;29(1):169-175. doi:10.1158/1055-9965.Epi-19-0781
  35. Shuai Y, Li C, Zhou X. The effect of metformin on gastric cancer in patients with type 2 diabetes: a systematic review and meta-analysis. Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico . Feb 14 2020;doi:10.1007/s12094-020-02304-y
  36. Dulskas A, Patasius A, Kaceniene A, Linkeviciute-Ulinskiene D, Zabuliene L, Smailyte G. A Cohort Study of Antihyperglycemic Medication Exposure and Gastric Cancer Risk. J Clin Med. Feb 5 2020;9(2)doi:10.3390/jcm9020435
  37. Danila E, Linkeviciute-Ulinskiene D, Zablockis R, Gruslys V, Cicenas S, Smailyte G. A Cohort Study of Exposure to Antihyperglycemic Therapy and Survival in Patients with Lung Cancer. International journal of environmental research and public health. Mar 7 2020;17(5)doi:10.3390/ijerph17051747
  38. Afzal MZ, Dragnev K, Sarwar T, Shirai K. Clinical outcomes in non-small-cell lung cancer patients receiving concurrent metformin and immune checkpoint inhibitors. Lung Cancer Manag. Oct 2019;8(2):Lmt11. doi:10.2217/lmt-2018-0016
  39. Kangwan N, Kim YJ, Han YM, Jeong M, Park JM, Hahm KB. Concerted actions of ameliorated colitis, aberrant crypt foci inhibition and 15-hydroxyprostaglandin dehydrogenase induction by sonic hedgehog inhibitor led to prevention of colitis-associated cancer. International journal of cancer Journal international du cancer. Mar 15 2016;138(6):1482-93. doi:10.1002/ijc.29892
  40. Wang Y, Liu X, Yan P, et al. Effect of metformin on the risk of prostate cancer in patients with type 2 diabetes by considering different confounding factors: a meta-analysis of observational studies. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation (ECP) . Jan 2020;29(1):42-52. doi:10.1097/cej.0000000000000514
  41. He K, Hu H, Ye S, Wang H, Cui R, Yi L. The effect of metformin therapy on incidence and prognosis in prostate cancer: A systematic review and meta-analysis. Sci Rep. Feb 18 2019;9(1):2218. doi:10.1038/s41598-018-38285-w
  42. Ghiasi B, Sarokhani D, Najafi F, Motedayen M, Dehkordi AH. The Relationship Between Prostate Cancer and Metformin Consumption: A Systematic Review and Meta-analysis Study. Curr Pharm Des. 2019;25(9):1021-1029. doi:10.2174/1381612825666190215123759
  43. Seliger C, Luber C, Gerken M, et al. Use of metformin and survival of patients with high-grade glioma. International journal of cancer Journal international du cancer. Jan 15 2019;144(2):273-280. doi:10.1002/ijc.31783
  44. Seliger C, Genbrugge E, Gorlia T, et al. Use of metformin and outcome of patients with newly diagnosed glioblastoma: Pooled analysis. International journal of cancer Journal international du cancer. Feb 1 2020;146(3):803-809. doi:10.1002/ijc.32337
  45. Wen Q, Zhao Z, Wen J, et al. The association between metformin therapy and risk of gynecological cancer in patients: Two meta-analyses. European journal of obstetrics, gynecology, and reproductive biology . Jun 2019;237:33-41. doi:10.1016/j.ejogrb.2019.03.029
  46. Tian J, Liang Y, Qu P. Antidiabetic Medications and the Risk of Endometrial Cancer in Patients. Gynecol Obstet Invest. 2019;84(5):455-462. doi:10.1159/000497202
  47. Chu D, Wu J, Wang K, et al. Effect of metformin use on the risk and prognosis of endometrial cancer: a systematic review and meta-analysis. BMC cancer. Apr 18 2018;18(1):438. doi:10.1186/s12885-018-4334-5
  48. Xie W, Li T, Yang J, et al. Metformin use and survival outcomes in endometrial cancer: a systematic review and meta-analysis. Oncotarget. Sep 22 2017;8(42):73079-73086. doi:10.18632/oncotarget.20388
  49. Tang YL, Zhu LY, Li Y, et al. Metformin Use Is Associated with Reduced Incidence and Improved Survival of Endometrial Cancer: A Meta-Analysis. Biomed Res Int. 2017;2017:5905384. doi:10.1155/2017/5905384
  50. Perez-Lopez FR, Pasupuleti V, Gianuzzi X, Palma-Ardiles G, Hernandez-Fernandez W, Hernandez AV. Systematic review and meta-analysis of the effect of metformin treatment on overall mortality rates in women with endometrial cancer and type 2 diabetes mellitus. Maturitas. Jul 2017;101:6-11. doi:10.1016/j.maturitas.2017.04.001
  51. Guo J, Xu K, An M, Zhao Y. Metformin and endometrial cancer survival: a quantitative synthesis of observational studies. Oncotarget. Sep 12 2017;8(39):66169-66177. doi:10.18632/oncotarget.19830
  52. Tseng CH. Metformin is associated with a lower risk of non-Hodgkin lymphoma in patients with type 2 diabetes. Diabetes Metab. Oct 2019;45(5):458-464. doi:10.1016/j.diabet.2019.05.002
  53. Ye X, Zhang G, Righolt C, et al. Metformin Is Not Associated with Incidence Risk of Non-Hodgkin Lymphomas among Diabetic Patients. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . May 2018;27(5):610-612. doi:10.1158/1055-9965.Epi-18-0012
  54. Wang Y, Maurer MJ, Larson MC, et al. Impact of metformin use on the outcomes of newly diagnosed diffuse large B-cell lymphoma and follicular lymphoma. Br J Haematol. Sep 2019;186(6):820-828. doi:10.1111/bjh.15997
  55. Alkhatib Y, Abdel Rahman Z, Kuriakose P. Clinical impact of metformin in diabetic diffuse large B-cell lymphoma patients: a case-control study. Leuk Lymphoma. May 2017;58(5):1130-1134. doi:10.1080/10428194.2016.1239822
  56. Boursi B, Mamtani R, Yang YX, Weiss BM. Impact of metformin on the progression of MGUS to multiple myeloma. Leuk Lymphoma. May 2017;58(5):1265-1267. doi:10.1080/10428194.2016.1236375
  57. Chang SH, Luo S, O'Brian KK, et al. Association between metformin use and progression of monoclonal gammopathy of undetermined significance to multiple myeloma in US veterans with diabetes mellitus: a population-based retrospective cohort study. Lancet Haematol. Jan 2015;2(1):e30-6. doi:10.1016/s2352-3026(14)00037-4
  58. Wu W, Merriman K, Nabaah A, et al. The association of diabetes and anti-diabetic medications with clinical outcomes in multiple myeloma. British journal of cancer. Jul 29 2014;111(3):628-36. doi:10.1038/bjc.2014.307
  59. Wang QL, Santoni G, Ness-Jensen E, Lagergren J, Xie SH. Association Between Metformin Use and Risk of Esophageal Squamous Cell Carcinoma in a Population-Based Cohort Study. The American journal of gastroenterology. Jan 2020;115(1):73-78. doi:10.14309/ajg.0000000000000478
  60. Tseng CH. Metformin and esophageal cancer risk in Taiwanese patients with type 2 diabetes mellitus. Oncotarget. Mar 21 2017;8(12):18802-18810. doi:10.18632/oncotarget.13390
  61. Wang Y, Fu T, Liu Y, Yang G, Yu C, Zhang ZJ. The association between metformin and head and neck cancer survival: a systematic review and meta-analysis of cohort studies. Curr Pharm Des. Feb 17 2020;doi:10.2174/1381612826666200218095310
  62. DeCensi A, Puntoni M, Guerrieri-Gonzaga A, et al. Effect of Metformin on Breast Ductal Carcinoma In Situ Proliferation in a Randomized Presurgical Trial. Cancer prevention research (Philadelphia, Pa). Oct 2015;8(10):888-94. doi:10.1158/1940-6207.capr-15-0048
  63. Martin-Castillo B, Pernas S, Dorca J, et al. A phase 2 trial of neoadjuvant metformin in combination with trastuzumab and chemotherapy in women with early HER2-positive breast cancer: the METTEN study. Oncotarget. Nov 2 2018;9(86):35687-35704. doi:10.18632/oncotarget.26286
  64. Lopez-Bonet E, Buxo M, Cuyas E, et al. Neoadjuvant Metformin Added to Systemic Therapy Decreases the Proliferative Capacity of Residual Breast Cancer. J Clin Med. Dec 11 2019;8(12)doi:10.3390/jcm8122180
  65. Rothermundt C, Hayoz S, Templeton AJ, et al. Metformin in chemotherapy-naive castration-resistant prostate cancer: a multicenter phase 2 trial (SAKK 08/09). Eur Urol. Sep 2014;66(3):468-74. doi:10.1016/j.eururo.2013.12.057
  66. Marrone KA, Zhou X, Forde PM, et al. A Randomized Phase II Study of Metformin plus Paclitaxel/Carboplatin/Bevacizumab in Patients with Chemotherapy-Naive Advanced or Metastatic Nonsquamous Non-Small Cell Lung Cancer. The oncologist. Feb 27 2018;doi:10.1634/theoncologist.2017-0465
  67. Arrieta O, Barron F, Padilla MS, et al. Effect of Metformin Plus Tyrosine Kinase Inhibitors Compared With Tyrosine Kinase Inhibitors Alone in Patients With Epidermal Growth Factor Receptor-Mutated Lung Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA oncology. Sep 5 2019:e192553. doi:10.1001/jamaoncol.2019.2553
  68. Ramos-Penafiel C, Olarte-Carrillo I, Ceron-Maldonado R, et al. Effect of metformin on the survival of patients with ALL who express high levels of the ABCB1 drug resistance gene. Journal of translational medicine. Sep 3 2018;16(1):245. doi:10.1186/s12967-018-1620-6
  69. Higurashi T, Hosono K, Takahashi H, et al. Metformin for chemoprevention of metachronous colorectal adenoma or polyps in post-polypectomy patients without diabetes: a multicentre double-blind, placebo-controlled, randomised phase 3 trial. The Lancet Oncology. Apr 2016;17(4):475-483. doi:10.1016/s1470-2045(15)00565-3
  70. Zheng Y, Zhu J, Zhang H, Liu Y, Sun H. Metformin plus first-line chemotherapy versus chemotherapy alone in the treatment of epithelial ovarian cancer: a prospective open-label pilot trial. Cancer chemotherapy and pharmacology. Dec 2019;84(6):1349-1357. doi:10.1007/s00280-019-03963-7
  71. Chen K, Li Y, Guo Z, Zeng Y, Zhang W, Wang H. Metformin: current clinical applications in nondiabetic patients with cancer. Aging (Albany NY). Feb 18 2020;12(4):3993-4009. doi:10.18632/aging.102787
  72. NIH. National Institutes of Health: US National Library of Medicine: MedlinePlus. Metformin. Available at https://medlineplus.gov/druginfo/meds/a696005.html. Last updated 05/21/2018. Accessed 05/31/2018. 2018;
  73. Schernthaner G, Schernthaner GH. The right place for metformin today. Diabetes Res Clin Pract. Jan 2020;159:107946. doi:10.1016/j.diabres.2019.107946
  74. Zhang Q, Li S, Li L, et al. Metformin Treatment and Homocysteine: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients. Dec 9 2016;8(12)doi:10.3390/nu8120798
  75. Krysiak R, Okopien B. The effect of metformin on androgen production in diabetic women with non-classic congenital adrenal hyperplasia. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association . Nov 2014;122(10):568-71. doi:10.1055/s-0034-1382048
  76. Al-Kuraishy HM, Al-Gareeb AI. Erectile Dysfunction and Low Sex Drive in Men with Type 2 DM: The Potential Role of Diabetic Pharmacotherapy. Journal of clinical and diagnostic research : JCDR. 2016;10(12):FC21-FC26. doi:10.7860/JCDR/2016/19971.8996
  77. Pasquali R, Gambineri A, Biscotti D, et al. Effect of long-term treatment with metformin added to hypocaloric diet on body composition, fat distribution, and androgen and insulin levels in abdominally obese women with and without the polycystic ovary syndrome. J Clin Endocrinol Metab. Aug 2000;85(8):2767-74. doi:10.1210/jcem.85.8.6738
  78. Akhter MS, Uppal P. Toxicity of Metformin and Hypoglycemic Therapies. Adv Chronic Kidney Dis. Jan 2020;27(1):18-30. doi:10.1053/j.ackd.2019.08.004
  79. Willemsen AE, Bredie SJ, Lobo CM, van der Vlugt MJ, Kramers C. [Choosing wisely when prescribing statins]. Nederlands tijdschrift voor geneeskunde. 2015;159:A8695. Verstandig kiezen bij het voorschrijven van statinen.
  80. Fatehi Hassanabad A. Current perspectives on statins as potential anti-cancer therapeutics: clinical outcomes and underlying molecular mechanisms. Transl Lung Cancer Res. Oct 2019;8(5):692-699. doi:10.21037/tlcr.2019.09.08
  81. Dutta A, Sharma-Walia N. Curbing Lipids: Impacts ON Cancer and Viral Infection. International journal of molecular sciences. Feb 2 2019;20(3)doi:10.3390/ijms20030644
  82. Chimento A, Casaburi I, Avena P, et al. Cholesterol and Its Metabolites in Tumor Growth: Therapeutic Potential of Statins in Cancer Treatment. Frontiers in endocrinology. 2018;9:807. doi:10.3389/fendo.2018.00807
  83. Allen SC, Mamotte CDS. Pleiotropic and Adverse Effects of Statins-Do Epigenetics Play a Role? The Journal of pharmacology and experimental therapeutics. Aug 2017;362(2):319-326. doi:10.1124/jpet.117.242081
  84. Iannelli F, Lombardi R, Milone MR, et al. Targeting Mevalonate Pathway in Cancer Treatment: Repurposing of Statins. Recent patents on anti-cancer drug discovery. 2018;13(2):184-200. doi:10.2174/1574892812666171129141211
  85. Hu YB, Hu ED, Fu RQ. Statin Use and Cancer Incidence in Patients with Type 2 Diabetes Mellitus: A Network Meta-Analysis. Gastroenterology research and practice. 2018;2018:8620682. doi:10.1155/2018/8620682
  86. Jeong GH, Lee KH, Kim JY, et al. Effect of Statin on Cancer Incidence: An Umbrella Systematic Review and Meta-Analysis. J Clin Med. Jun 8 2019;8(6)doi:10.3390/jcm8060819
  87. Emilsson L, Garcia-Albeniz X, Logan RW, Caniglia EC, Kalager M, Hernan MA. Examining Bias in Studies of Statin Treatment and Survival in Patients With Cancer. JAMA oncology. Jan 1 2018;4(1):63-70. doi:10.1001/jamaoncol.2017.2752
  88. He Y, Li X, Gasevic D, et al. Statins and Multiple Noncardiovascular Outcomes: Umbrella Review of Meta-analyses of Observational Studies and Randomized Controlled Trials. Ann Intern Med. Oct 16 2018;169(8):543-553. doi:10.7326/m18-0808
  89. Kim J, You NY, Lee JW, Kim Y, Kim YW, Kang HT. Inverse Association Between Statin Use and Overall Cancer Incidence in Individuals With Hypercholesterolemia, Based on the Korean Health Insurance Service Between 2002 and 2015. Asia Pac J Public Health. Mar 2019;31(2):136-146. doi:10.1177/1010539519830235
  90. Mei Z, Liang M, Li L, Zhang Y, Wang Q, Yang W. Effects of statins on cancer mortality and progression: A systematic review and meta-analysis of 95 cohorts including 1,111,407 individuals. International journal of cancer Journal international du cancer. Mar 01 2017;140(5):1068-1081. doi:10.1002/ijc.30526
  91. Craveiro NS, Silva Lopes B, Tomas L, et al. L-TRUST: Long-term risk of cancer in patients under statins therapy. A systematic review and meta-analysis. Pharmacoepidemiology and drug safety. Nov 2019;28(11):1431-1439. doi:10.1002/pds.4895
  92. Galicia-Garcia U, Jebari S, Larrea-Sebal A, et al. Statin Treatment-Induced Development of Type 2 Diabetes: From Clinical Evidence to Mechanistic Insights. International journal of molecular sciences. Jul 2 2020;21(13)doi:10.3390/ijms21134725
  93. Tran KT, McMenamin UC, Coleman HG, et al. Statin use and risk of liver cancer: Evidence from two population-based studies. International journal of cancer Journal international du cancer. Mar 1 2020;146(5):1250-1260. doi:10.1002/ijc.32426
  94. Islam MM, Poly TN, Walther BA, Yang HC, Jack Li YC. Statin Use and the Risk of Hepatocellular Carcinoma: A Meta-Analysis of Observational Studies. Cancers. Mar 13 2020;12(3)doi:10.3390/cancers12030671
  95. Kim G, Jang SY, Nam CM, Kang ES. Statin use and the risk of hepatocellular carcinoma in patients at high risk: A nationwide nested case-control study. J Hepatol. Mar 2018;68(3):476-484. doi:10.1016/j.jhep.2017.10.018
  96. Thrift AP, Natarajan Y, Liu Y, El-Serag HB. Statin Use After Diagnosis of Hepatocellular Carcinoma Is Associated With Decreased Mortality. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association . Sep 2019;17(10):2117-2125.e3. doi:10.1016/j.cgh.2018.12.046
  97. Simon TG, Duberg AS, Aleman S, et al. Lipophilic Statins and Risk for Hepatocellular Carcinoma and Death in Patients With Chronic Viral Hepatitis: Results From a Nationwide Swedish Population. Ann Intern Med. Sep 3 2019;171(5):318-327. doi:10.7326/m18-2753
  98. Thomas T, Loke Y, Beales ILP. Systematic Review and Meta-analysis: Use of Statins Is Associated with a Reduced Incidence of Oesophageal Adenocarcinoma. J Gastrointest Cancer. Dec 2018;49(4):442-454. doi:10.1007/s12029-017-9983-0
  99. Deng HY, Lan X, Zheng X, et al. The association between statin use and survival of esophageal cancer patients: A systematic review and meta-analysis. Medicine. Jul 2019;98(29):e16480. doi:10.1097/md.0000000000016480
  100. You HS, You N, Lee JW, Lim HJ, Kim J, Kang HT. Inverse Association between Statin Use and Stomach Cancer Incidence in Individuals with Hypercholesterolemia, from the 2002-2015 NHIS-HEALS Data. International journal of environmental research and public health. Feb 7 2020;17(3)doi:10.3390/ijerph17031054
  101. Cheung KS, Chan EW, Wong AYS, et al. Statins Were Associated with a Reduced Gastric Cancer Risk in Patients with Eradicated Helicobacter Pylori Infection: A Territory-Wide Propensity Score Matched Study. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Feb 2020;29(2):493-499. doi:10.1158/1055-9965.Epi-19-1044
  102. Chung H, Kim HJ, Jung HC, Lee SK, Kim SG. Statins and metachronous recurrence after endoscopic resection of early gastric cancer: a nationwide Korean cohort study. Gastric cancer : official journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association . Jan 24 2020;doi:10.1007/s10120-020-01041-z
  103. Spence AD, Busby J, Hughes CM, Johnston BT, Coleman HG, Cardwell CR. Statin use and survival in patients with gastric cancer in two independent population-based cohorts. Pharmacoepidemiology and drug safety. Apr 2019;28(4):460-470. doi:10.1002/pds.4688
  104. Hamada T, Khalaf N, Yuan C, et al. Statin use and pancreatic cancer risk in two prospective cohort studies. Journal of gastroenterology. Aug 2018;53(8):959-966. doi:10.1007/s00535-018-1430-x
  105. Zhang Y, Liang M, Sun C, et al. Statin Use and Risk of Pancreatic Cancer: An Updated Meta-analysis of 26 Studies. Pancreas. Feb 2019;48(2):142-150. doi:10.1097/mpa.0000000000001226
  106. Archibugi L, Arcidiacono PG, Capurso G. Statin use is associated to a reduced risk of pancreatic cancer: A meta-analysis. Dig Liver Dis. Jan 2019;51(1):28-37. doi:10.1016/j.dld.2018.09.007
  107. Bang UC, Watanabe T, Bendtsen F. The relationship between the use of statins and mortality, severity, and pancreatic cancer in Danish patients with chronic pancreatitis. European journal of gastroenterology & hepatology. Mar 2018;30(3):346-351. doi:10.1097/meg.0000000000001060
  108. Choi JH, Lee SH, Huh G, et al. The association between use of statin or aspirin and pancreatic ductal adenocarcinoma: A nested case-control study in a Korean nationwide cohort. Cancer Med. Dec 2019;8(17):7419-7430. doi:10.1002/cam4.2617
  109. Kirkegard J, Lund JL, Mortensen FV, Cronin-Fenton D. Statins and pancreatic cancer risk in patients with chronic pancreatitis: A Danish nationwide population-based cohort study. International journal of cancer Journal international du cancer. Feb 1 2020;146(3):610-616. doi:10.1002/ijc.32264
  110. Tamburrino D, Crippa S, Partelli S, et al. Statin use improves survival in patients with pancreatic ductal adenocarcinoma: A meta-analysis. Dig Liver Dis. Feb 26 2020;doi:10.1016/j.dld.2020.01.008
  111. Abdel-Rahman O. Statin treatment and outcomes of metastatic pancreatic cancer: a pooled analysis of two phase III studies. Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico . Jun 2019;21(6):810-816. doi:10.1007/s12094-018-1992-3
  112. Hamada T, Khalaf N, Yuan C, et al. Prediagnosis Use of Statins Associates With Increased Survival Times of Patients With Pancreatic Cancer. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association . Aug 2018;16(8):1300-1306.e3. doi:10.1016/j.cgh.2018.02.022
  113. Mondul AM, Joshu CE, Barber JR, et al. Longer-term Lipid-lowering Drug Use and Risk of Incident and Fatal Prostate Cancer in Black and White Men in the ARIC Study. Cancer prevention research (Philadelphia, Pa). Dec 2018;11(12):779-788. doi:10.1158/1940-6207.Capr-17-0396
  114. Tan P, Zhang C, Wei SY, et al. Effect of statins type on incident prostate cancer risk: a meta-analysis and systematic review. Asian journal of andrology. Nov-Dec 2017;19(6):666-671. doi:10.4103/1008-682x.190327
  115. Van Rompay MI, Solomon KR, Nickel JC, Ranganathan G, Kantoff PW, McKinlay JB. Prostate cancer incidence and mortality among men using statins and non-statin lipid-lowering medications. European journal of cancer (Oxford, England : 1990). May 2019;112:118-126. doi:10.1016/j.ejca.2018.11.033
  116. Dawe DE, Ye X, Czaykowski P, et al. The effect of statin use on the incidence of prostate cancer: A population-based nested case-control study. International journal of cancer Journal international du cancer. Jul 1 2018;143(1):190-198. doi:10.1002/ijc.31295
  117. Meijer D, van Moorselaar RJA, Vis AN, Bijnsdorp IV. Prostate Cancer Development Is Not Affected by Statin Use in Patients with Elevated PSA Levels. Cancers. Jul 7 2019;11(7)doi:10.3390/cancers11070953
  118. Jayalath VH, Nayan M, Finelli A, et al. Statin use and time to progression in men on active surveillance for prostate cancer. Prostate cancer and prostatic diseases. Nov 2018;21(4):509-515. doi:10.1038/s41391-018-0053-x
  119. Anderson-Carter I, Posielski N, Liou JI, et al. The impact of statins in combination with androgen deprivation therapyin patients with advanced prostate cancer: A large observational study. Urologic oncology. Feb 2019;37(2):130-137. doi:10.1016/j.urolonc.2018.11.017
  120. Wu SY, Fang SC, Shih HJ, Wen YC, Shao YJ. Mortality associated with statins in men with advanced prostate cancer treated with androgen deprivation therapy. European journal of cancer (Oxford, England : 1990). May 2019;112:109-117. doi:10.1016/j.ejca.2018.11.032
  121. Murtola TJ, Peltomaa AI, Talala K, et al. Statin Use and Prostate Cancer Survival in the Finnish Randomized Study of Screening for Prostate Cancer. European urology focus. Apr 2017;3(2-3):212-220. doi:10.1016/j.euf.2016.05.004
  122. Larsen SB, Dehlendorff C, Skriver C, et al. Postdiagnosis Statin Use and Mortality in Danish Patients With Prostate Cancer. J Clin Oncol. Oct 10 2017;35(29):3290-3297. doi:10.1200/jco.2016.71.8981
  123. Allott EH, Ebot EM, Stopsack KH, et al. Statin Use Is Associated with Lower Risk of PTEN-Null and Lethal Prostate Cancer. Clin Cancer Res. Mar 1 2020;26(5):1086-1093. doi:10.1158/1078-0432.Ccr-19-2853
  124. Wang K, Gerke TA, Chen X, Prosperi M. Association of statin use with risk of Gleason score-specific prostate cancer: A hospital-based cohort study. Cancer Med. Dec 2019;8(17):7399-7407. doi:10.1002/cam4.2500
  125. Van Wyhe RD, Rahal OM, Woodward WA. Effect of statins on breast cancer recurrence and mortality: a review. Breast cancer (Dove Medical Press). 2017;9:559-565. doi:10.2147/bctt.S148080
  126. Islam MM, Yang HC, Nguyen PA, et al. Exploring association between statin use and breast cancer risk: an updated meta-analysis. Archives of gynecology and obstetrics. Dec 2017;296(6):1043-1053. doi:10.1007/s00404-017-4533-3
  127. Liu B, Yi Z, Guan X, Zeng YX, Ma F. The relationship between statins and breast cancer prognosis varies by statin type and exposure time: a meta-analysis. Breast cancer research and treatment. Jul 2017;164(1):1-11. doi:10.1007/s10549-017-4246-0
  128. Li YR, Ro V, Steel L, et al. Impact of long-term lipid-lowering therapy on clinical outcomes in breast cancer. Breast cancer research and treatment. Aug 2019;176(3):669-677. doi:10.1007/s10549-019-05267-z
  129. Borgquist S, Broberg P, Tojjar J, Olsson H. Statin use and breast cancer survival - a Swedish nationwide study. BMC cancer. Jan 11 2019;19(1):54. doi:10.1186/s12885-018-5263-z
  130. Langballe R, Cronin-Fenton D, Dehlendorff C, et al. Statin use and risk of contralateral breast cancer: a nationwide cohort study. British journal of cancer. Nov 2018;119(10):1297-1305. doi:10.1038/s41416-018-0252-1
  131. Hosio M, Urpilainen E, Marttila M, et al. Association of antidiabetic medication and statins with breast cancer incidence in women with type 2 diabetes. Breast cancer research and treatment. Jun 2019;175(3):741-748. doi:10.1007/s10549-019-05185-0
  132. Kwon YJ, You NY, Lee JW, Kim J, Kang HT. High Receipt of Statins Reduces the Risk of Lung Cancer in Current Smokers With Hypercholesterolemia: The National Health Insurance Service-Health Screening Cohort. Clinical lung cancer. Mar 2019;20(2):e177-e185. doi:10.1016/j.cllc.2018.11.009
  133. Liu JC, Yang TY, Hsu YP, et al. Statins dose-dependently exert a chemopreventive effect against lung cancer in COPD patients: a population-based cohort study. Oncotarget. Sep 13 2016;7(37):59618-59629. doi:10.18632/oncotarget.11162
  134. Xia DK, Hu ZG, Tian YF, Zeng FJ. Statin use and prognosis of lung cancer: a systematic review and meta-analysis of observational studies and randomized controlled trials. Drug design, development and therapy. 2019;13:405-422. doi:10.2147/dddt.S187690
  135. Chen Y, Li X, Zhang R, Xia Y, Shao Z, Mei Z. Effects of statin exposure and lung cancer survival: A meta-analysis of observational studies. Pharmacological research : the official journal of the Italian Pharmacological Society . Mar 2019;141:357-365. doi:10.1016/j.phrs.2019.01.016
  136. Oh TK, Kim K, Jheon S, et al. Impact of Statin Use on Recurrence or Survival After Surgical Curative Resection of Non-Small Cell Lung Cancer. Cancer control : journal of the Moffitt Cancer Center. Jan-Mar 2018;25(1):1073274818778000. doi:10.1177/1073274818778000
  137. Shah SC, Glass J, Giustino G, et al. Statin Exposure Is Not Associated with Reduced Prevalence of Colorectal Neoplasia in Patients with Inflammatory Bowel Disease. Gut and liver. Jan 15 2019;13(1):54-61. doi:10.5009/gnl18178
  138. Mak JWY, So J, Tang W, et al. Cancer risk and chemoprevention in Chinese inflammatory bowel disease patients: a population-based cohort study. Scandinavian journal of gastroenterology. Mar 2 2020:1-8. doi:10.1080/00365521.2020.1731760
  139. Renman D, Lundberg E, Gunnarsson U, Strigård K. Statin consumption as a risk factor for developing colorectal cancer: a retrospective case study. World journal of surgical oncology. Dec 16 2017;15(1):222. doi:10.1186/s12957-017-1287-0
  140. Lee JW, You NY, Kim Y, Kim Y, Kim J, Kang HT. Statin use and site-specific risk of colorectal cancer in individuals with hypercholesterolemia from the National Health Insurance Service-National Health Screening Cohort (NHIS-HEALS). Nutr Metab Cardiovasc Dis. Jul 2019;29(7):701-709. doi:10.1016/j.numecd.2019.04.002
  141. Bowles EJA, Yu O, Ziebell R, et al. Cardiovascular medication use and risks of colon cancer recurrences and additional cancer events: a cohort study. BMC cancer. Mar 27 2019;19(1):270. doi:10.1186/s12885-019-5493-8
  142. Lash TL, Riis AH, Ostenfeld EB, et al. Associations of Statin Use With Colorectal Cancer Recurrence and Mortality in a Danish Cohort. American journal of epidemiology. Sep 15 2017;186(6):679-687. doi:10.1093/aje/kww245
  143. Dobrzycka M, Spychalski P, Lachinski AJ, Kobiela P, Jedrusik P, Kobiela J. Statins and Colorectal Cancer - A Systematic Review. Experimental and clinical endocrinology & diabetes : official journal, German Society of Endocrinology [and] German Diabetes Association . Aug 27 2018;doi:10.1055/a-0668-5692
  144. Gray RT, Coleman HG, Hughes C, Murray LJ, Cardwell CR. Statin use and survival in colorectal cancer: Results from a population-based cohort study and an updated systematic review and meta-analysis. Cancer epidemiology. Dec 2016;45:71-81. doi:10.1016/j.canep.2016.10.004
  145. Voorneveld PW, Reimers MS, Bastiaannet E, et al. Statin Use After Diagnosis of Colon Cancer and Patient Survival. Gastroenterology. Aug 2017;153(2):470-479.e4. doi:10.1053/j.gastro.2017.05.011
  146. Wang Y, Ren F, Song Z, Chen P, Liu S, Ouyang L. Statin use and the risk of ovarian and endometrial cancers: a meta-analysis. BMC cancer. Jul 24 2019;19(1):730. doi:10.1186/s12885-019-5954-0
  147. Desai P, Wallace R, Anderson ML, et al. An analysis of the association between statin use and risk of endometrial and ovarian cancers in the Women's Health Initiative. Gynecol Oncol. Mar 2018;148(3):540-546. doi:10.1016/j.ygyno.2018.01.006
  148. Arima R, Marttila M, Hautakoski A, et al. Antidiabetic medication, statins and the risk of endometrioid endometrial cancer in patients with type 2 diabetes. Gynecol Oncol. Sep 2017;146(3):636-641. doi:10.1016/j.ygyno.2017.06.011
  149. Arima R, Marttila M, Hautakoski A, et al. Antidiabetic Medication, Statins and the Risk and Prognosis of Non-endometrioid Endometrial Cancer in Women with Type 2 Diabetes. Anticancer research. Jul 2018;38(7):4169-4178. doi:10.21873/anticanres.12710
  150. Sperling CD, Verdoodt F, Kjaer Hansen M, Dehlendorff C, Friis S, Kjaer SK. Statin use and mortality among endometrial cancer patients: a Danish nationwide cohort study. International journal of cancer Journal international du cancer. Dec 1 2018;143(11):2668-2676. doi:10.1002/ijc.31625
  151. Segev Y, Gemer O, Helpman L, et al. An Israeli Gynecologic Oncology Group study of statin use and endometrial cancer prognosis. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics . Jan 2020;148(1):79-86. doi:10.1002/ijgo.12981
  152. Urpilainen E, Marttila M, Hautakoski A, et al. The role of metformin and statins in the incidence of epithelial ovarian cancer in type 2 diabetes: a cohort and nested case-control study. BJOG : an international journal of obstetrics and gynaecology. Jul 2018;125(8):1001-1008. doi:10.1111/1471-0528.15151
  153. Akinwunmi B, Vitonis AF, Titus L, Terry KL, Cramer DW. Statin therapy and association with ovarian cancer risk in the New England Case Control (NEC) study. International journal of cancer Journal international du cancer. Mar 1 2019;144(5):991-1000. doi:10.1002/ijc.31758
  154. Harding BN, Delaney JA, Urban RR, Weiss NS. Use of Statin Medications Following Diagnosis in Relation to Survival among Women with Ovarian Cancer. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Jul 2019;28(7):1127-1133. doi:10.1158/1055-9965.Epi-18-1194
  155. Li X, Zhou J. Impact of postdiagnostic statin use on ovarian cancer mortality: A systematic review and meta-analysis of observational studies. Br J Clin Pharmacol. Jun 2018;84(6):1109-1120. doi:10.1111/bcp.13559
  156. Kao LT, Hung SH, Kao PF, Liu JC, Lin HC. Inverse association between statin use and head and neck cancer: Population-based case-control study in Han population. Head & neck. May 2019;41(5):1193-1198. doi:10.1002/hed.25501
  157. Gupta A, Stokes W, Eguchi M, et al. Statin use associated with improved overall and cancer specific survival in patients with head and neck cancer. Oral oncology. Mar 2019;90:54-66. doi:10.1016/j.oraloncology.2019.01.019
  158. Chou YC, Lin CH, Wong CS, Chou WY, Chang JY, Sun CA. Statin use and the risk of renal cell carcinoma: national cohort study. Journal of investigative medicine : the official publication of the American Federation for Clinical Research . Mar 2020;68(3):776-781. doi:10.1136/jim-2019-001209
  159. Nayan M, Punjani N, Juurlink DN, et al. Statin use and kidney cancer survival outcomes: A systematic review and meta-analysis. Cancer treatment reviews. Jan 2017;52:105-116. doi:10.1016/j.ctrv.2016.11.009
  160. Neumann E, Klaiber P, Freitag K, et al. Assessment of concomitant non-oncologic medication in patients with surgically treated renal cell carcinoma: impact on prognosis, cell-cycle progression and proliferation. Journal of cancer research and clinical oncology. Jul 2019;145(7):1835-1843. doi:10.1007/s00432-019-02914-2
  161. Righolt CH, Zhang G, Ye X, et al. Statin Use and Chronic Lymphocytic Leukemia Incidence: A Nested Case-Control Study in Manitoba, Canada. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Sep 2019;28(9):1495-1501. doi:10.1158/1055-9965.Epi-19-0107
  162. Desai P, Wallace R, Anderson ML, et al. An analysis of the effect of statins on the risk of Non-Hodgkin's Lymphoma in the Women's Health Initiative cohort. Cancer Med. May 2018;7(5):2121-2130. doi:10.1002/cam4.1368
  163. Epstein MM, Divine G, Chao CR, et al. Statin use and risk of multiple myeloma: An analysis from the cancer research network. International journal of cancer Journal international du cancer. Aug 1 2017;141(3):480-487. doi:10.1002/ijc.30745
  164. Cote DJ, Rosner BA, Smith-Warner SA, Egan KM, Stampfer MJ. Statin use, hyperlipidemia, and risk of glioma. European journal of epidemiology. Nov 2019;34(11):997-1011. doi:10.1007/s10654-019-00565-8
  165. Xie Y, Lu Q, Lenahan C, Yang S, Zhou D, Qi X. Whether statin use improves the survival of patients with glioblastoma?: A meta-analysis. Medicine. Feb 2020;99(9):e18997. doi:10.1097/md.0000000000018997
  166. Seliger C, Schaertl J, Gerken M, et al. Use of statins or NSAIDs and survival of patients with high-grade glioma. PLoS One. 2018;13(12):e0207858. doi:10.1371/journal.pone.0207858
  167. Ahmadi M, Amiri S, Pecic S, et al. Pleiotropic effects of statins: A focus on cancer. Biochim Biophys Acta Mol Basis Dis. Dec 1 2020;1866(12):165968. doi:10.1016/j.bbadis.2020.165968
  168. Farooqi MAM, Malhotra N, Mukherjee SD, et al. Statin therapy in the treatment of active cancer: A systematic review and meta-analysis of randomized controlled trials. PLoS One. 2018;13(12):e0209486. doi:10.1371/journal.pone.0209486
  169. Jang HJ, Kim HS, Kim JH, Lee J. The Effect of Statin Added to Systemic Anticancer Therapy: A Meta-Analysis of Randomized, Controlled Trials. J Clin Med. Oct 4 2018;7(10)doi:10.3390/jcm7100325
  170. Kim MK, Myung SK, Tran BT, Park B. Statins and risk of cancer: A meta-analysis of randomized, double-blind, placebo-controlled trials. Indian journal of cancer. Apr-Jun 2017;54(2):470-477. doi:10.4103/ijc.IJC_214_17
  171. Toth PP, Patti AM, Giglio RV, et al. Management of Statin Intolerance in 2018: Still More Questions Than Answers. American Journal of Cardiovascular Drugs. 2018/06/01 2018;18(3):157-173. doi:10.1007/s40256-017-0259-7
  172. Muller AL, Freed DH. Basic and Clinical Observations of Mevalonate Depletion on the Mevalonate Signaling Pathway. Current molecular pharmacology. 2017;10(1):6-12. doi:10.2174/1874467209666160112125805
  173. Garrido-Maraver J, Cordero MD, Oropesa-Avila M, et al. Clinical applications of coenzyme Q10. Frontiers in bioscience (Landmark edition). Jan 1 2014;19:619-33.
  174. Banach M, Serban C, Ursoniu S, et al. Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group. Statin therapy and plasma coenzyme Q10 concentrations—a systematic review and meta-analysis of placebo-controlled trials. Pharmacological research : the official journal of the Italian Pharmacological Society . 2015;99:329-336.
  175. NIH: National Cancer Institute. Aspirin to reduce cancer risk. https://www.cancer.gov/about-cancer/causes-prevention/research/aspirin-cancer-risk
  176. Dulai PS, Singh S, Marquez E, et al. Chemoprevention of colorectal cancer in individuals with previous colorectal neoplasia: systematic review and network meta-analysis. BMJ (Clinical research ed). Dec 05 2016;355:i6188. doi:10.1136/bmj.i6188
  177. Smith CJ, Dorsey TH, Tang W, Jordan SV, Loffredo CA, Ambs S. Aspirin Use Reduces the Risk of Aggressive Prostate Cancer and Disease Recurrence in African-American Men. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Jun 2017;26(6):845-853. doi:10.1158/1055-9965.epi-16-1027
  178. Hybiak J, Broniarek I, Kiryczynski G, et al. Aspirin and its pleiotropic application. European journal of pharmacology. Jan 5 2020;866:172762. doi:10.1016/j.ejphar.2019.172762
  179. Todoric J, Antonucci L, Karin M. Targeting Inflammation in Cancer Prevention and Therapy. Cancer prevention research (Philadelphia, Pa). Dec 2016;9(12):895-905. doi:10.1158/1940-6207.Capr-16-0209
  180. Moris D, Kontos M, Spartalis E, Fentiman IS. The Role of NSAIDs in Breast Cancer Prevention and Relapse: Current Evidence and Future Perspectives. Breast care (Basel, Switzerland). Oct 2016;11(5):339-344. doi:10.1159/000452315
  181. Riesenberg BP, Ansa-Addo EA, Gutierrez J, Timmers CD, Liu B, Li Z. Cutting Edge: Targeting Thrombocytes to Rewire Anticancer Immunity in the Tumor Microenvironment and Potentiate Efficacy of PD-1 Blockade. J Immunol. Sep 1 2019;203(5):1105-1110. doi:10.4049/jimmunol.1900594
  182. Kanikarla-Marie P, Lam M, Sorokin AV, Overman MJ, Kopetz S, Menter DG. Platelet Metabolism and Other Targeted Drugs; Potential Impact on Immunotherapy. Frontiers in oncology. 2018;8:107. doi:10.3389/fonc.2018.00107
  183. Bashir AIJ, Kankipati CS, Jones S, et al. A novel mechanism for the anticancer activity of aspirin and salicylates. International journal of oncology. Apr 2019;54(4):1256-1270. doi:10.3892/ijo.2019.4701
  184. Zhang X, Feng Y, Liu X, et al. Beyond a chemopreventive reagent, aspirin is a master regulator of the hallmarks of cancer. Journal of cancer research and clinical oncology. Jun 2019;145(6):1387-1403. doi:10.1007/s00432-019-02902-6
  185. Qiao Y, Yang T, Gan Y, et al. Associations between aspirin use and the risk of cancers: a meta-analysis of observational studies. BMC cancer. Mar 13 2018;18(1):288. doi:10.1186/s12885-018-4156-5
  186. Tsoi KKF, Ho JMW, Chan FCH, Sung JJY. Long-term use of low-dose aspirin for cancer prevention: A 10-year population cohort study in Hong Kong. International journal of cancer Journal international du cancer. Jul 1 2019;145(1):267-273. doi:10.1002/ijc.32083
  187. Zheng SL, Roddick AJ. Association of Aspirin Use for Primary Prevention With Cardiovascular Events and Bleeding Events: A Systematic Review and Meta-analysis. JAMA. 2019;321(3):277-287. doi:10.1001/jama.2018.20578
  188. Loomans-Kropp HA, Pinsky P, Cao Y, Chan AT, Umar A. Association of Aspirin Use With Mortality Risk Among Older Adult Participants in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. JAMA Netw Open. Dec 2 2019;2(12):e1916729. doi:10.1001/jamanetworkopen.2019.16729
  189. Ajrouche A, De Rycke Y, Dalichampt M, et al. Reduced risk of cancer among low-dose aspirin users: Data from French health care databases. Pharmacoepidemiology and drug safety. Sep 2019;28(9):1258-1266. doi:10.1002/pds.4870
  190. Frere C, Lejeune M, Kubicek P, Faille D, Marjanovic Z. Antiplatelet Agents for Cancer Prevention: Current Evidences and Continuing Controversies. Cancers. Oct 24 2019;11(11)doi:10.3390/cancers11111639
  191. McNeil JJ, Nelson MR, Woods RL, et al. Effect of Aspirin on All-Cause Mortality in the Healthy Elderly. The New England journal of medicine. Oct 18 2018;379(16):1519-1528. doi:10.1056/NEJMoa1803955
  192. Bowman L, Mafham M, Wallendszus K, et al. Effects of Aspirin for Primary Prevention in Persons with Diabetes Mellitus. The New England journal of medicine. Oct 18 2018;379(16):1529-1539. doi:10.1056/NEJMoa1804988
  193. Okada S, Morimoto T, Ogawa H, et al. Effect of Aspirin on Cancer Chemoprevention in Japanese Patients With Type 2 Diabetes: 10-Year Observational Follow-up of a Randomized Controlled Trial. Diabetes Care. Aug 2018;41(8):1757-1764. doi:10.2337/dc18-0368
  194. Wang S, Yu Y, Ryan PM, et al. Association of aspirin therapy with risk of hepatocellular carcinoma: A systematic review and dose-response analysis of cohort studies with 2.5 million participants. Pharmacological research : the official journal of the Italian Pharmacological Society . Jan 2020;151:104585. doi:10.1016/j.phrs.2019.104585
  195. Shin S, Lee SH, Lee M, et al. Aspirin and the risk of hepatocellular carcinoma development in patients with alcoholic cirrhosis. Medicine. Feb 2020;99(9):e19008. doi:10.1097/md.0000000000019008
  196. Simon TG, Duberg AS, Aleman S, Chung RT, Chan AT, Ludvigsson JF. Association of Aspirin with Hepatocellular Carcinoma and Liver-Related Mortality. The New England journal of medicine. Mar 12 2020;382(11):1018-1028. doi:10.1056/NEJMoa1912035
  197. Lee TY, Hsu YC, Tseng HC, et al. Association of Daily Aspirin Therapy With Risk of Hepatocellular Carcinoma in Patients With Chronic Hepatitis B. JAMA Intern Med. May 1 2019;179(5):633-640. doi:10.1001/jamainternmed.2018.8342
  198. Young SH, Chau GY, Lee IC, et al. Aspirin is associated with low recurrent risk in hepatitis B virus-related hepatocellular carcinoma patients after curative resection. Journal of the Formosan Medical Association = Taiwan yi zhi. Jan 2020;119(1 Pt 2):218-229. doi:10.1016/j.jfma.2019.04.018
  199. Trabert B, Poole EM, White E, et al. Analgesic Use and Ovarian Cancer Risk: An Analysis in the Ovarian Cancer Cohort Consortium. Journal of the National Cancer Institute. Feb 1 2019;111(2):137-145. doi:10.1093/jnci/djy100
  200. Zhang D, Bai B, Xi Y, Wang T, Zhao Y. Is aspirin use associated with a decreased risk of ovarian cancer? A systematic review and meta-analysis of observational studies with dose-response analysis. Gynecol Oncol. Aug 2016;142(2):368-77. doi:10.1016/j.ygyno.2016.04.543
  201. Barnard ME, Poole EM, Curhan GC, et al. Association of Analgesic Use With Risk of Ovarian Cancer in the Nurses' Health Studies. JAMA oncology. Dec 1 2018;4(12):1675-1682. doi:10.1001/jamaoncol.2018.4149
  202. Merritt MA, Rice MS, Barnard ME, et al. Pre-diagnosis and post-diagnosis use of common analgesics and ovarian cancer prognosis (NHS/NHSII): a cohort study. The Lancet Oncology. Aug 2018;19(8):1107-1116. doi:10.1016/s1470-2045(18)30373-5
  203. Verdoodt F, Kjaer SK, Dehlendorff C, Friis S. Aspirin use and ovarian cancer mortality in a Danish nationwide cohort study. British journal of cancer. Feb 20 2018;118(4):611-615. doi:10.1038/bjc.2017.449
  204. Wield AM, Walsh CS, Rimel BJ, Cass I, Karlan BY, Li AJ. Aspirin use correlates with survival in women with clear cell ovarian cancer. Gynecol Oncol Rep. Aug 2018;25:78-81. doi:10.1016/j.gore.2018.06.004
  205. Nagle CM, Ibiebele TI, DeFazio A, Protani MM, Webb PM. Aspirin, nonaspirin nonsteroidal anti-inflammatory drugs, acetaminophen and ovarian cancer survival. Cancer epidemiology. Apr 2015;39(2):196-9. doi:10.1016/j.canep.2014.12.010
  206. Lapumnuaypol K, Tiu A, Thongprayoon C, et al. Effects of aspirin and non-steroidal anti-inflammatory drugs on the risk of cholangiocarcinoma: a meta-analysis. Qjm. Jun 1 2019;112(6):421-427. doi:10.1093/qjmed/hcz039
  207. Xiong J, Xu W, Bian J, et al. Aspirin use is associated with a reduced risk of cholangiocarcinoma: a systematic review and meta-analysis. Cancer management and research. 2018;10:4095-4104. doi:10.2147/cmar.S173197
  208. Jackson SS, Pfeiffer RM, Liu Z, et al. Association Between Aspirin Use and Biliary Tract Cancer Survival. JAMA oncology. Oct 17 2019;doi:10.1001/jamaoncol.2019.4328
  209. Huang XZ, Chen Y, Wu J, et al. Aspirin and non-steroidal anti-inflammatory drugs use reduce gastric cancer risk: A dose-response meta-analysis. Oncotarget. Jan 17 2017;8(3):4781-4795. doi:10.18632/oncotarget.13591
  210. Kong P, Wu R, Liu X, et al. The Effects of Anti-inflammatory Drug Treatment in Gastric Cancer Prevention: an Update of a Meta-analysis. Journal of Cancer. 2016;7(15):2247-2257. doi:10.7150/jca.16524
  211. Brusselaers N, Lagergren J. Maintenance use of non-steroidal anti-inflammatory drugs and risk of gastrointestinal cancer in a nationwide population-based cohort study in Sweden. BMJ open. Jul 7 2018;8(7):e021869. doi:10.1136/bmjopen-2018-021869
  212. Kim YI, Kim SY, Kim JH, et al. Long-Term Low-Dose Aspirin Use Reduces Gastric Cancer Incidence: A Nationwide Cohort Study. Cancer research and treatment : official journal of Korean Cancer Association . Apr 2016;48(2):798-805. doi:10.4143/crt.2015.117
  213. Wang Y, Shen C, Ge J, Duan H. Regular aspirin use and stomach cancer risk in China. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology . Jun 2015;41(6):801-4. doi:10.1016/j.ejso.2015.02.006
  214. Kim MH, Chang J, Kim WJ, Banerjee S, Park SM. Cumulative Dose Threshold for the Chemopreventive Effect of Aspirin Against Gastric Cancer. The American journal of gastroenterology. Jun 2018;113(6):845-854. doi:10.1038/s41395-018-0097-5
  215. Spence AD, Busby J, Johnston BT, et al. Low-Dose Aspirin Use Does Not Increase Survival in 2 Independent Population-Based Cohorts of Patients With Esophageal or Gastric Cancer. Gastroenterology. Mar 2018;154(4):849-860.e1. doi:10.1053/j.gastro.2017.10.044
  216. Clarke CA, Canchola AJ, Moy LM, et al. Regular and low-dose aspirin, other non-steroidal anti-inflammatory medications and prospective risk of HER2-defined breast cancer: the California Teachers Study. Breast Cancer Res. May 1 2017;19(1):52. doi:10.1186/s13058-017-0840-7
  217. Lu L, Shi L, Zeng J, Wen Z. Aspirin as a potential modality for the chemoprevention of breast cancer: A dose-response meta-analysis of cohort studies from 857,831 participants. Oncotarget. Jun 20 2017;8(25):40389-40401. doi:10.18632/oncotarget.16315
  218. Kehm RD, Hopper JL, John EM, et al. Regular use of aspirin and other non-steroidal anti-inflammatory drugs and breast cancer risk for women at familial or genetic risk: a cohort study. Breast Cancer Res. Apr 18 2019;21(1):52. doi:10.1186/s13058-019-1135-y
  219. Bens A, Friis S, Dehlendorff C, et al. Low-dose aspirin use and risk of contralateral breast cancer: a Danish nationwide cohort study. Preventive medicine. Nov 2018;116:186-193. doi:10.1016/j.ypmed.2018.09.015
  220. Elwood PC, Pickering JE, Morgan G, et al. Systematic review update of observational studies further supports aspirin role in cancer treatment: Time to share evidence and decision-making with patients? PLoS One . 2018;13(9):e0203957. doi:10.1371/journal.pone.0203957
  221. Frisk G, Ekberg S, Lidbrink E, et al. No association between low-dose aspirin use and breast cancer outcomes overall: a Swedish population-based study. Breast Cancer Res. Nov 20 2018;20(1):142. doi:10.1186/s13058-018-1065-0
  222. Wang T, McCullough LE, White AJ, et al. Prediagnosis aspirin use, DNA methylation, and mortality after breast cancer: A population-based study. Cancer. Nov 1 2019;125(21):3836-3844. doi:10.1002/cncr.32364
  223. Sauer CM, Myran DT, Costentin CE, et al. Effect of long term aspirin use on the incidence of prostate cancer: A systematic review and meta-analysis. Critical reviews in oncology/hematology. Dec 2018;132:66-75. doi:10.1016/j.critrevonc.2018.09.013
  224. Prause LW, Manka L, Millan C, et al. Influence of regular aspirin intake on PSA values, prostate cancer incidence and overall survival in a prospective screening trial (ERSPC Aarau). World J Urol. Dec 21 2019;doi:10.1007/s00345-019-03054-5
  225. Hurwitz LM, Joshu CE, Barber JR, et al. Aspirin and Non-Aspirin NSAID Use and Prostate Cancer Incidence, Mortality, and Case Fatality in the Atherosclerosis Risk in Communities Study. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Mar 2019;28(3):563-569. doi:10.1158/1055-9965.Epi-18-0965
  226. Downer MK, Allard CB, Preston MA, et al. Aspirin Use and Lethal Prostate Cancer in the Health Professionals Follow-up Study. Eur Urol Oncol . Mar 2019;2(2):126-134. doi:10.1016/j.euo.2018.07.002
  227. Skriver C, Dehlendorff C, Borre M, et al. Use of Low-Dose Aspirin and Mortality After Prostate Cancer Diagnosis: A Nationwide Cohort Study. Ann Intern Med. Apr 2 2019;170(7):443-452. doi:10.7326/m17-3085
  228. Zhou J, Xia S, Li T, Liu R. Could aspirin be a lifesaver for prostate cancer patients in prostate cancer-specific mortality?: an update systematic review and meta-analysis. BMC cancer. Dec 5 2019;19(1):1186. doi:10.1186/s12885-019-6415-5
  229. Fan LL, Xie CP, Wu YM, Gu XJ, Chen YH, Wang YJ. Aspirin Exposure and Mortality Risk among Prostate Cancer Patients: A Systematic Review and Meta-Analysis. Biomed Res Int. 2019;2019:9379602. doi:10.1155/2019/9379602
  230. Zhang YP, Wan YD, Sun YL, Li J, Zhu RT. Aspirin might reduce the incidence of pancreatic cancer: A meta-analysis of observational studies. Sci Rep. Oct 21 2015;5:15460. doi:10.1038/srep15460
  231. Sun J, Li Y, Liu L, Jiang Z, Liu G. Aspirin use and pancreatic cancer risk: A systematic review of observational studies. Medicine. Dec 2019;98(51):e18033. doi:10.1097/md.0000000000018033
  232. Risch HA, Lu L, Streicher SA, et al. Aspirin Use and Reduced Risk of Pancreatic Cancer. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Jan 2017;26(1):68-74. doi:10.1158/1055-9965.Epi-16-0508
  233. Khalaf N, Yuan C, Hamada T, et al. Regular Use of Aspirin or Non-Aspirin Nonsteroidal Anti-Inflammatory Drugs Is Not Associated With Risk of Incident Pancreatic Cancer in Two Large Cohort Studies. Gastroenterology. Apr 2018;154(5):1380-1390.e5. doi:10.1053/j.gastro.2017.12.001
  234. Kim MH, Park SM, Yun YH, Hwang IC. Aspirin Does Not Prevent Pancreatic Cancer in a Large Asian Cohort. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Apr 2019;28(4):826-828. doi:10.1158/1055-9965.Epi-18-1325
  235. Jiang HY, Huang TB, Xu L, et al. Aspirin use and lung cancer risk: a possible relationship? Evidence from an updated meta-analysis. PLoS One. 2015;10(4):e0122962. doi:10.1371/journal.pone.0122962
  236. Hochmuth F, Jochem M, Schlattmann P. Meta-analysis of aspirin use and risk of lung cancer shows notable results. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation (ECP) . Jul 2016;25(4):259-68. doi:10.1097/cej.0000000000000176
  237. Ye S, Lee M, Lee D, Ha EH, Chun EM. Association of Long-term Use of Low-Dose Aspirin as Chemoprevention With Risk of Lung Cancer. JAMA Netw Open. Mar 1 2019;2(3):e190185. doi:10.1001/jamanetworkopen.2019.0185
  238. Erickson P, Gardner LD, Loffredo CA, et al. Racial and Ethnic Differences in the Relationship between Aspirin Use and Non-Small Cell Lung Cancer Risk and Survival. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Dec 2018;27(12):1518-1526. doi:10.1158/1055-9965.Epi-18-0366
  239. Sandelin M, Mindus S, Thuresson M, et al. Factors associated with lung cancer in COPD patients. Int J Chron Obstruct Pulmon Dis. 2018;13:1833-1839. doi:10.2147/copd.S162484
  240. Mc Menamin UC, Cardwell CR, Hughes CM, Murray LM. Low-dose aspirin and survival from lung cancer: a population-based cohort study. BMC cancer. Nov 17 2015;15:911. doi:10.1186/s12885-015-1910-9
  241. Maddison P. Effects of aspirin on small-cell lung cancer mortality and metastatic presentation. Lung cancer (Amsterdam, Netherlands). Apr 2017;106:67-69. doi:10.1016/j.lungcan.2017.01.018
  242. Lohinai Z, Dome P, Szilagyi Z, et al. From Bench to Bedside: Attempt to Evaluate Repositioning of Drugs in the Treatment of Metastatic Small Cell Lung Cancer (SCLC). PLoS One. 2016;11(1):e0144797. doi:10.1371/journal.pone.0144797
  243. Webb PM, Na R, Weiderpass E, et al. Use of aspirin, other nonsteroidal anti-inflammatory drugs and acetaminophen and risk of endometrial cancer: the Epidemiology of Endometrial Cancer Consortium. Ann Oncol. Feb 1 2019;30(2):310-316. doi:10.1093/annonc/mdy541
  244. Takiuchi T, Blake EA, Matsuo K, Sood AK, Brasky TM. Aspirin use and endometrial cancer risk and survival. Gynecol Oncol. Jan 2018;148(1):222-232. doi:10.1016/j.ygyno.2017.10.026
  245. Verdoodt F, Kjaer SK, Friis S. Influence of aspirin and non-aspirin NSAID use on ovarian and endometrial cancer: Summary of epidemiologic evidence of cancer risk and prognosis. Maturitas. Jun 2017;100:1-7. doi:10.1016/j.maturitas.2017.03.001
  246. Zhang D, Bai B, Xi Y, Zhao Y. Can Aspirin Reduce the Risk of Endometrial Cancer?: A Systematic Review and Meta-analysis of Observational Studies. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society . Jul 2016;26(6):1111-20. doi:10.1097/igc.0000000000000731
  247. Verdoodt F, Friis S, Dehlendorff C, Albieri V, Kjaer SK. Non-steroidal anti-inflammatory drug use and risk of endometrial cancer: A systematic review and meta-analysis of observational studies. Gynecol Oncol. Feb 2016;140(2):352-8. doi:10.1016/j.ygyno.2015.12.009
  248. Matsuo K, Cahoon SS, Yoshihara K, et al. Association of Low-Dose Aspirin and Survival of Women With Endometrial Cancer. Obstet Gynecol. Jul 2016;128(1):127-37. doi:10.1097/aog.0000000000001491
  249. Sperling CD, Verdoodt F, Aalborg GL, Dehlendorff C, Friis S, Kjaer SK. Low-dose aspirin use and endometrial cancer mortality-a Danish nationwide cohort study. Int J Epidemiol. Dec 17 2019;doi:10.1093/ije/dyz253
  250. Amirian ES, Ostrom QT, Armstrong GN, et al. Aspirin, NSAIDs, and Glioma Risk: Original Data from the Glioma International Case-Control Study and a Meta-analysis. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Mar 2019;28(3):555-562. doi:10.1158/1055-9965.Epi-18-0702
  251. Lumley CJ, Kaffenberger TM, Desale S, et al. Post-diagnosis aspirin use and survival in veterans with head and neck cancer. Head & neck. May 2019;41(5):1220-1226. doi:10.1002/hed.25518
  252. Lin YS, Yeh CC, Huang SF, et al. Aspirin associated with risk reduction of secondary primary cancer for patients with head and neck cancer: A population-based analysis. PLoS One. 2018;13(8):e0199014. doi:10.1371/journal.pone.0199014
  253. Chang H, Tao YL, Ye WJ, Xiao WW, Xia YF, Gao YH. Regular aspirin intake and prognosis of TxN2-3M0 nasopharyngeal carcinoma: A cohort study based on propensity score matching. Oral oncology. Feb 11 2020;103:104589. doi:10.1016/j.oraloncology.2020.104589
  254. Araujo JL, Altorki NK, Sonett JR, et al. Prediagnosis aspirin use and outcomes in a prospective cohort of esophageal cancer patients. Therapeutic advances in gastroenterology. Nov 2016;9(6):806-814. doi:10.1177/1756283x16657985
  255. McNeil JJ, Nelson MR, Woods RL, et al. Effect of Aspirin on All-Cause Mortality in the Healthy Elderly. New England Journal of Medicine. 2018;379(16):1519-1528. doi:10.1056/NEJMoa1803955
  256. McNeil JJ, Gibbs P, Orchard SG, et al. Effect of aspirin on cancer incidence and mortality in older adults. JNCI: Journal of the National Cancer Institute. 2020;doi:10.1093/jnci/djaa114
  257. Hull MA, Sprange K, Hepburn T, et al. Eicosapentaenoic acid and aspirin, alone and in combination, for the prevention of colorectal adenomas (seAFOod Polyp Prevention trial): a multicentre, randomised, double-blind, placebo-controlled, 2 x 2 factorial trial. Lancet. Dec 15 2018;392(10164):2583-2594. doi:10.1016/s0140-6736(18)31775-6
  258. Coyle C, Cafferty FH, Rowley S, et al. ADD-ASPIRIN: A phase III, double-blind, placebo controlled, randomised trial assessing the effects of aspirin on disease recurrence and survival after primary therapy in common non-metastatic solid tumours. Contemporary clinical trials. Nov 2016;51:56-64. doi:10.1016/j.cct.2016.10.004
  259. Miyamoto K, Takashima A, Mizusawa J, et al. Efficacy of aspirin for stage III colorectal cancer: a randomized double-blind placebo-controlled trial (JCOG1503C, EPISODE-III trial). Japanese journal of clinical oncology. Oct 1 2019;49(10):985-990. doi:10.1093/jjco/hyz106
  260. Petrera M, Paleari L, Clavarezza M, et al. The ASAMET trial: a randomized, phase II, double-blind, placebo-controlled, multicenter, 2 x 2 factorial biomarker study of tertiary prevention with low-dose aspirin and metformin in stage I-III colorectal cancer patients. BMC cancer. Dec 4 2018;18(1):1210. doi:10.1186/s12885-018-5126-7
  261. Michel P, Boige V, Andre T, et al. Aspirin versus placebo in stage III or high-risk stage II colon cancer with PIK3CA mutation: A French randomised double-blind phase III trial (PRODIGE 50-ASPIK). Dig Liver Dis. Mar 2018;50(3):305-307. doi:10.1016/j.dld.2017.12.023
  262. Haykal T, Barbarawi M, Zayed Y, et al. Safety and efficacy of aspirin for primary prevention of cancer: a meta-analysis of randomized controlled trials. Journal of cancer research and clinical oncology. Jul 2019;145(7):1795-1809. doi:10.1007/s00432-019-02932-0
  263. NIH. National Institutes of Health: US National Library of Medicine: Medline Plus: Diclofenac. Available at https://medlineplus.gov/druginfo/meds/a689002.html#why. Last updated 04/15/2020. Accessed 08/03/2020. 2020;
  264. Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
  265. Pandey SK, Yadav S, Goel Y, Temre MK, Singh VK, Singh SM. Molecular docking of anti-inflammatory drug diclofenac with metabolic targets: Potential applications in cancer therapeutics. J Theor Biol. Mar 21 2019;465:117-125. doi:10.1016/j.jtbi.2019.01.020
  266. Thomas GJ, Herranz P, Cruz SB, Parodi A. Treatment of actinic keratosis through inhibition of cyclooxygenase-2: Potential mechanism of action of diclofenac sodium 3% in hyaluronic acid 2.5. Dermatologic therapy. May 2019;32(3):e12800. doi:10.1111/dth.12800
  267. Arisan ED, Akar RO, Rencuzogullari O, et al. The molecular targets of diclofenac differs from ibuprofen to induce apoptosis and epithelial mesenchymal transition due to alternation on oxidative stress management p53 independently in PC3 prostate cancer cells. Prostate Int. Dec 2019;7(4):156-165. doi:10.1016/j.prnil.2019.09.003
  268. Morgan AGM, Babu D, Michail K, Siraki AG. An evaluation of myeloperoxidase-mediated bio-activation of NSAIDs in promyelocytic leukemia (HL-60) cells for potential cytotoxic selectivity. Toxicol Lett. Oct 5 2017;280:48-56. doi:10.1016/j.toxlet.2017.07.894
  269. Gerthofer V, Kreutz M, Renner K, et al. Combined Modulation of Tumor Metabolism by Metformin and Diclofenac in Glioma. International journal of molecular sciences. Aug 31 2018;19(9)doi:10.3390/ijms19092586
  270. Duval AP, Troquier L, de Souza Silva O, Demartines N, Dormond O. Diclofenac Potentiates Sorafenib-Based Treatments of Hepatocellular Carcinoma by Enhancing Oxidative Stress. Cancers. Sep 27 2019;11(10)doi:10.3390/cancers11101453
  271. Intini FP, Zajac J, Novohradsky V, et al. Novel Antitumor Platinum(II) Conjugates Containing the Nonsteroidal Anti-inflammatory Agent Diclofenac: Synthesis and Dual Mechanisms of Antiproliferative Effects. Inorg Chem. Feb 6 2017;56(3):1483-1497. doi:10.1021/acs.inorgchem.6b02553
  272. Will OM, Purcz N, Chalaris A, et al. Increased survival rate by local release of diclofenac in a murine model of recurrent oral carcinoma. International journal of nanomedicine. 2016;11:5311-5321. doi:10.2147/ijn.S109199
  273. Schack A, Fransgaard T, Klein MF, Gögenur I. Perioperative Use of Nonsteroidal Anti-inflammatory Drugs Decreases the Risk of Recurrence of Cancer After Colorectal Resection: A Cohort Study Based on Prospective Data. Annals of surgical oncology. Nov 2019;26(12):3826-3837. doi:10.1245/s10434-019-07600-8
  274. Hung CH, Lin YC, Chang YH, et al. The effect of NSAIDs exposure on breast cancer risk in female patients with autoimmune diseases. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation (ECP) . Sep 2019;28(5):428-434. doi:10.1097/cej.0000000000000476
  275. Cronin-Fenton DP, Heide-Jørgensen U, Ahern TP, et al. Low-dose Aspirin, Nonsteroidal Anti-inflammatory Drugs, Selective COX-2 Inhibitors and Breast Cancer Recurrence. Epidemiology (Cambridge, Mass). Jul 2016;27(4):586-93. doi:10.1097/ede.0000000000000480
  276. Desmedt C, Demicheli R, Fornili M, et al. Potential Benefit of Intra-operative Administration of Ketorolac on Breast Cancer Recurrence According to the Patient's Body Mass Index. J Natl Cancer Inst. Oct 1 2018;110(10):1115-1122. doi:10.1093/jnci/djy042
  277. Javor S, Cozzani E, Parodi A. Topical treatment of actinic keratosis with 3.0% diclofenac in 2.5% hyaluronan gel: review of the literature about the cumulative evidence of its efficacy and safety. G Ital Dermatol Venereol. Jun 2016;151(3):275-80.
  278. Wolf JE, Jr., Taylor JR, Tschen E, Kang S. Topical 3.0% diclofenac in 2.5% hyaluronan gel in the treatment of actinic keratoses. Int J Dermatol. Nov 2001;40(11):709-13. doi:10.1046/j.1365-4362.2001.01324.x
  279. Rivers JK, Arlette J, Shear N, Guenther L, Carey W, Poulin Y. Topical treatment of actinic keratoses with 3.0% diclofenac in 2.5% hyaluronan gel. Br J Dermatol. Jan 2002;146(1):94-100. doi:10.1046/j.1365-2133.2002.04561.x
  280. Steeb T, Wessely A, Harlaß M, et al. A Systematic Review and Meta-Analysis of Interventions for Actinic Keratosis from Post-Marketing Surveillance Trials. J Clin Med. Jul 15 2020;9(7)doi:10.3390/jcm9072253
  281. Gollnick H, Dirschka T, Ostendorf R, Kerl H, Kunstfeld R. Long-term clinical outcomes of imiquimod 5% cream vs. diclofenac 3% gel for actinic keratosis on the face or scalp: a pooled analysis of two randomized controlled trials. J Eur Acad Dermatol Venereol. Jan 2020;34(1):82-89. doi:10.1111/jdv.15868
  282. Wong RSY. Role of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) in Cancer Prevention and Cancer Promotion. Advances in pharmacological sciences. 2019;2019:3418975. doi:10.1155/2019/3418975
  283. Faustino-Rocha AI, Ferreira R, Gama A, Oliveira PA, Ginja M. Antihistamines as promising drugs in cancer therapy. Life Sci. Mar 1 2017;172:27-41. doi:10.1016/j.lfs.2016.12.008
  284. Ellegaard AM, Dehlendorff C, Vind AC, et al. Repurposing Cationic Amphiphilic Antihistamines for Cancer Treatment. EBioMedicine. Jul 2016;9:130-139. doi:10.1016/j.ebiom.2016.06.013
  285. Soule BP, Simone NL, DeGraff WG, Choudhuri R, Cook JA, Mitchell JB. Loratadine dysregulates cell cycle progression and enhances the effect of radiation in human tumor cell lines. Radiation oncology (London, England). Feb 3 2010;5:8. doi:10.1186/1748-717x-5-8
  286. Chen T, Hu Y, Liu B, et al. Combining thioridazine and loratadine for the treatment of gastrointestinal tumor. Oncology letters. Oct 2017;14(4):4573-4580. doi:10.3892/ol.2017.6815
  287. Verdoodt F, Dehlendorff C, Jaattela M, et al. Antihistamines and ovarian cancer survival: nationwide cohort study and in vitro cell viability assay. Journal of the National Cancer Institute. Nov 5 2019;doi:10.1093/jnci/djz217
  288. Bens A, Dehlendorff C, Friis S, et al. The role of H1 antihistamines in contralateral breast cancer: a Danish nationwide cohort study. British journal of cancer. Feb 17 2020;doi:10.1038/s41416-020-0747-4
  289. Kirshner JJ, McDonald MC, 3rd, Kruter F, et al. NOLAN: a randomized, phase 2 study to estimate the effect of prophylactic naproxen or loratadine vs no prophylactic treatment on bone pain in patients with early-stage breast cancer receiving chemotherapy and pegfilgrastim. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer . Apr 2018;26(4):1323-1334. doi:10.1007/s00520-017-3959-2
  290. Loratadine: Drug information (Lexicomp). UpToDate. Updated 2020. Accessed 09/29/2020, https://www.uptodate.com/contents/loratadine-drug-information?search=loratadine&source=panel_search_result&selectedTitle=1~66&usage_type=panel&kp_tab=drug_general&display_rank=1#F6699703
  291. Jafarzadeh A, Nemati M, Khorramdelazad H, Hassan ZM. Immunomodulatory properties of cimetidine: Its therapeutic potentials for treatment of immune-related diseases. International immunopharmacology. May 2019;70:156-166. doi:10.1016/j.intimp.2019.02.026
  292. Takahashi HK, Watanabe T, Yokoyama A, et al. Cimetidine induces interleukin-18 production through H2-agonist activity in monocytes. Mol Pharmacol. Aug 2006;70(2):450-3. doi:10.1124/mol.106.025890
  293. Kubota T, Fujiwara H, Ueda Y, et al. Cimetidine modulates the antigen presenting capacity of dendritic cells from colorectal cancer patients. British journal of cancer. Apr 22 2002;86(8):1257-61. doi:10.1038/sj.bjc.6600233
  294. Furuta T, Sabit H, Dong Y, et al. Biological basis and clinical study of glycogen synthase kinase- 3beta-targeted therapy by drug repositioning for glioblastoma. Oncotarget. Apr 4 2017;8(14):22811-22824. doi:10.18632/oncotarget.15206
  295. Ali AH, Hale L, Yalamanchili B, et al. The Effect of Perioperative Cimetidine Administration on Time to Colorectal Cancer Recurrence. American journal of therapeutics. Jul/Aug 2018;25(4):e405-e411. doi:10.1097/mjt.0000000000000547
  296. Tonnesen H, Knigge U, Bulow S, et al. Effect of cimetidine on survival after gastric cancer. Lancet. Oct 29 1988;2(8618):990-2.
  297. Burtin C, Noirot C, Scheinmann P, Galoppin L, Sabolovic D, Bernard P. Clinical improvement in advanced cancer disease after treatment combining histamine and H2-antihistaminics (ranitidine or cimetidine). European journal of cancer & clinical oncology. Feb 1988;24(2):161-7. doi:10.1016/0277-5379(88)90247-7
  298. Langman MJ, Dunn JA, Whiting JL, et al. Prospective, double-blind, placebo-controlled randomized trial of cimetidine in gastric cancer. British Stomach Cancer Group. British journal of cancer. Dec 1999;81(8):1356-62. doi:10.1038/sj.bjc.6690457
  299. Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP. Repurposing drugs in oncology (ReDO)-cimetidine as an anti-cancer agent. Ecancermedicalscience. 2014;8:485. doi:10.3332/ecancer.2014.485
  300. Liu FR, Jiang CG, Li YS, Li JB, Li F. Cimetidine inhibits the adhesion of gastric cancer cells expressing high levels of sialyl Lewis x in human vascular endothelial cells by blocking E-selectin expression. Int J Mol Med. Apr 2011;27(4):537-44. doi:10.3892/ijmm.2011.618
  301. Matsumoto S, Imaeda Y, Umemoto S, Kobayashi K, Suzuki H, Okamoto T. Cimetidine increases survival of colorectal cancer patients with high levels of sialyl Lewis-X and sialyl Lewis-A epitope expression on tumour cells. British journal of cancer. Jan 21 2002;86(2):161-7. doi:10.1038/sj.bjc.6600048
  302. NIH. National Institutes of Health: US National Library of Medicine: Medline Plus. Cimetidine. Available at https://medlineplus.gov/druginfo/meds/a682256.html. Last updated 04/23/2018. Accessed 05/22/2018. 2018;
  303. Fumagalli C, Maurizi N, Marchionni N, Fornasari D. beta-blockers: Their new life from hypertension to cancer and migraine. Pharmacological research : the official journal of the Italian Pharmacological Society . Jan 2020;151:104587. doi:10.1016/j.phrs.2019.104587
  304. Shin KJ, Lee YJ, Yang YR, et al. Molecular Mechanisms Underlying Psychological Stress and Cancer. Curr Pharm Des. 2016;22(16):2389-402.
  305. Inigo-Marco I, Alonso MM. Destress and do not suppress: targeting adrenergic signaling in tumor immunosuppression. J Clin Invest. Dec 2 2019;129(12):5086-5088. doi:10.1172/jci133115
  306. Krizanova O, Babula P, Pacak K. Stress, catecholaminergic system and cancer. Stress (Amsterdam, Netherlands). Jul 2016;19(4):419-28. doi:10.1080/10253890.2016.1203415
  307. Ma Y, Bai F, Qin F, et al. Beta-blockers for the primary prevention of anthracycline-induced cardiotoxicity: a meta-analysis of randomized controlled trials. BMC pharmacology & toxicology. Apr 25 2019;20(1):18. doi:10.1186/s40360-019-0298-6
  308. Fiala O, Ostasov P, Sorejs O, et al. Incidental Use of Beta-Blockers Is Associated with Outcome of Metastatic Colorectal Cancer Patients Treated with Bevacizumab-Based Therapy: A Single-Institution Retrospective Analysis of 514 Patients. Cancers. Nov 25 2019;11(12)doi:10.3390/cancers11121856
  309. Chang PY, Chung CH, Chang WC, et al. The effect of propranolol on the prognosis of hepatocellular carcinoma: A nationwide population-based study. PLoS One. 2019;14(5):e0216828. doi:10.1371/journal.pone.0216828
  310. Yap A, Lopez-Olivo MA, Dubowitz J, et al. Effect of beta-blockers on cancer recurrence and survival: a meta-analysis of epidemiological and perioperative studies. British journal of anaesthesia. Jul 2018;121(1):45-57. doi:10.1016/j.bja.2018.03.024
  311. Udumyan R, Montgomery S, Fang F, et al. Beta-Blocker Use and Lung Cancer Mortality in a Nationwide Cohort Study of Patients with Primary Non-Small Cell Lung Cancer. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Jan 2020;29(1):119-126. doi:10.1158/1055-9965.Epi-19-0710
  312. Coelho M, Squizzato A, Cassina N, Marino F, Ribeiro LV, Cosentino M. Effect of beta-blockers on survival of lung cancer patients: a systematic review and meta-analysis. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation (ECP) . Oct 11 2019;doi:10.1097/cej.0000000000000544
  313. Siltari A, Murtola TJ, Talala K, Taari K, Tammela TLJ, Auvinen A. Antihypertensive drugs and prostate cancer risk in a Finnish population-based cohort. Scand J Urol. Oct - Dec 2018;52(5-6):321-327. doi:10.1080/21681805.2018.1559882
  314. Kim M-S, Han KD, Kwon SY. Pre-diagnostic beta-blocker use and head- and neck cancer risk: A nationwide population-based case-control study. Medicine. 2019;98(24):e16047-e16047. doi:10.1097/MD.0000000000016047
  315. Kim MS, Han KD, Kwon SY. Pre-diagnostic beta-blocker use and head- and neck cancer risk: A nationwide population-based case-control study. Medicine. Jun 2019;98(24):e16047. doi:10.1097/md.0000000000016047
  316. Sanni OB, Mc Menamin UC, Cardwell CR, Sharp L, Murray LJ, Coleman HG. Commonly used medications and endometrial cancer survival: a population-based cohort study. British journal of cancer. Jul 25 2017;117(3):432-438. doi:10.1038/bjc.2017.207
  317. Harding BN, Delaney JA, Urban RR, Weiss NS. Post-diagnosis use of antihypertensive medications and the risk of death from ovarian cancer. Gynecol Oncol. Aug 2019;154(2):426-431. doi:10.1016/j.ygyno.2019.05.030
  318. Baek MH, Kim DY, Kim SO, Kim YJ, Park YH. Impact of beta blockers on survival outcomes in ovarian cancer: a nationwide population-based cohort study. Journal of gynecologic oncology. Nov 2018;29(6):e82. doi:10.3802/jgo.2018.29.e82
  319. Couttenier A, Lacroix O, Silversmit G, Vaes E, De Schutter H, Robert A. Beta-blocker use and mortality following ovarian cancer diagnosis: a population-based study. Cancer epidemiology. Oct 2019;62:101579. doi:10.1016/j.canep.2019.101579
  320. Phadke S, Clamon G. Beta blockade as adjunctive breast cancer therapy: A review. Critical reviews in oncology/hematology. Jun 2019;138:173-177. doi:10.1016/j.critrevonc.2019.04.006
  321. Hiller JG, Cole SW, Crone EM, et al. Preoperative beta-Blockade with Propranolol Reduces Biomarkers of Metastasis in Breast Cancer: A Phase II Randomized Trial. Clin Cancer Res. Nov 21 2019;doi:10.1158/1078-0432.Ccr-19-2641
  322. Souza VBd, Silva EN, Ribeiro ML, Martins WdA. Hypertension in patients with cancer. Arquivos brasileiros de cardiologia. 2015;104(3):246-252. doi:10.5935/abc.20150011
  323. Haldar R, Ricon-Becker I, Radin A, et al. Perioperative COX2 and β-adrenergic blockade improves biomarkers of tumor metastasis, immunity, and inflammation in colorectal cancer: A randomized controlled trial. Cancer. 2020;126(17):3991-4001. doi:10.1002/cncr.32950
  324. Ahl R, Matthiessen P, Fang X, et al. Effect of beta-blocker therapy on early mortality after emergency colonic cancer surgery. The British journal of surgery. Mar 2019;106(4):477-483. doi:10.1002/bjs.10988
  325. Ahl R, Matthiessen P, Cao Y, Sjolin G, Ljungqvist O, Mohseni S. The Relationship Between Severe Complications, Beta-Blocker Therapy and Long-Term Survival Following Emergency Surgery for Colon Cancer. World journal of surgery. Oct 2019;43(10):2527-2535. doi:10.1007/s00268-019-05058-z
  326. Knight JM, Rizzo JD, Hari P, et al. Propranolol inhibits molecular risk markers in HCT recipients: a phase 2 randomized controlled biomarker trial. Blood Advances. 2020;4(3):467-476. doi:10.1182/bloodadvances.2019000765
  327. Na Z, Qiao X, Hao X, et al. The effects of beta-blocker use on cancer prognosis: a meta-analysis based on 319,006 patients. OncoTargets and therapy. 2018;11:4913-4944. doi:10.2147/ott.S167422
  328. Pantziarka P, Bouche G, Sukhatme V, Meheus L, Rooman I, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-Propranolol as an anti-cancer agent. Ecancermedicalscience. 2016;10:680. doi:10.3332/ecancer.2016.680
  329. Lipska K, Gumieniczek A, Filip AA. Anticonvulsant valproic acid and other short-chain fatty acids as novel anticancer therapeutics: Possibilities and challenges. Acta pharmaceutica (Zagreb, Croatia). Sep 1 2020;70(3):291-301. doi:10.2478/acph-2020-0021
  330. Wawruszak A, Gumbarewicz E, Okon E, et al. Histone deacetylase inhibitors reinforce the phenotypical markers of breast epithelial or mesenchymal cancer cells but inhibit their migratory properties. Cancer management and research. 2019;11:8345-8358. doi:10.2147/cmar.S210029
  331. Luo Y, Wang H, Zhao X, et al. Valproic acid causes radiosensitivity of breast cancer cells via disrupting the DNA repair pathway. Toxicol Res (Camb). May 1 2016;5(3):859-870. doi:10.1039/c5tx00476d
  332. Terranova-Barberio M, Roca MS, Zotti AI, et al. Valproic acid potentiates the anticancer activity of capecitabine in vitro and in vivo in breast cancer models via induction of thymidine phosphorylase expression. Oncotarget. Feb 16 2016;7(7):7715-31. doi:10.18632/oncotarget.6802
  333. Kwiecinska P, Tauboll E, Grzyb E, Fiedor E, Ptak A, Gregoraszczuk EL. Valproic Acid as a Promising Co-Treatment With Paclitaxel and Doxorubicin in Different Ovarian Carcinoma Cell Lines. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society . Nov 2016;26(9):1546-1556. doi:10.1097/igc.0000000000000814
  334. Iwahashi S, Ishibashi H, Utsunomiya T, et al. Effect of histone deacetylase inhibitor in combination with 5-fluorouracil on pancreas cancer and cholangiocarcinoma cell lines. The journal of medical investigation : JMI. Feb 2011;58(1-2):106-9. doi:10.2152/jmi.58.106
  335. Sun G, Mackey LV, Coy DH, Yu CY, Sun L. The Histone Deacetylase Inhibitor Vaproic Acid Induces Cell Growth Arrest in Hepatocellular Carcinoma Cells via Suppressing Notch Signaling. Journal of Cancer. 2015;6(10):996-1004. doi:10.7150/jca.12135
  336. Tsaur I, Hudak L, Makarevic J, et al. Intensified antineoplastic effect by combining an HDAC-inhibitor, an mTOR-inhibitor and low dosed interferon alpha in prostate cancer cells. Journal of cellular and molecular medicine. Aug 2015;19(8):1795-804. doi:10.1111/jcmm.12583
  337. Raffoux E, Cras A, Recher C, et al. Phase 2 clinical trial of 5-azacitidine, valproic acid, and all-trans retinoic acid in patients with high-risk acute myeloid leukemia or myelodysplastic syndrome. Oncotarget. May 2010;1(1):34-42. doi:10.18632/oncotarget.100518
  338. Iwahashi S, Utsunomiya T, Imura S, et al. Effects of valproic acid in combination with S-1 on advanced pancreatobiliary tract cancers: clinical study phases I/II. Anticancer research. Sep 2014;34(9):5187-91.
  339. Mohammed TA, Holen KD, Jaskula-Sztul R, et al. A pilot phase II study of valproic acid for treatment of low-grade neuroendocrine carcinoma. The oncologist. 2011;16(6):835-43. doi:10.1634/theoncologist.2011-0031
  340. Falchook GS, Fu S, Naing A, et al. Methylation and histone deacetylase inhibition in combination with platinum treatment in patients with advanced malignancies. Investigational new drugs. Oct 2013;31(5):1192-200. doi:10.1007/s10637-013-0003-3
  341. Avallone A, Piccirillo MC, Di Gennaro E, et al. Randomized phase II study of valproic acid in combination with bevacizumab and oxaliplatin/fluoropyrimidine regimens in patients with RAS-mutated metastatic colorectal cancer: the REVOLUTION study protocol. Therapeutic advances in medical oncology. 2020;12:1758835920929589. doi:10.1177/1758835920929589
  342. Fushida S, Kinoshita J, Kaji M, et al. Paclitaxel plus valproic acid versus paclitaxel alone as second- or third-line therapy for advanced gastric cancer: a randomized Phase II trial. Drug design, development and therapy. 2016;10:2353-8. doi:10.2147/dddt.S110425
  343. Chu BF, Karpenko MJ, Liu Z, et al. Phase I study of 5-aza-2'-deoxycytidine in combination with valproic acid in non-small-cell lung cancer. Cancer chemotherapy and pharmacology. Jan 2013;71(1):115-21. doi:10.1007/s00280-012-1986-8
  344. Tataranni T, Piccoli C. Dichloroacetate (DCA) and Cancer: An Overview towards Clinical Applications. Oxid Med Cell Longev. 2019;2019:8201079. doi:10.1155/2019/8201079
  345. Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacological research : the official journal of the Italian Pharmacological Society . Dec 2019;150:104511. doi:10.1016/j.phrs.2019.104511
  346. Kankotia S, Stacpoole PW. Dichloroacetate and cancer: new home for an orphan drug? Biochim Biophys Acta. Dec 2014;1846(2):617-29. doi:10.1016/j.bbcan.2014.08.005
  347. Salamon S, Podbregar E, Kubatka P, et al. Glucose Metabolism in Cancer and Ischemia: Possible Therapeutic Consequences of the Warburg Effect. Nutrition and cancer. Feb-Mar 2017;69(2):177-183. doi:10.1080/01635581.2017.1263751
  348. Stakisaitis D, Jukneviciene M, Damanskiene E, Valanciute A, Balnyte I, Alonso MM. The Importance of Gender-Related Anticancer Research on Mitochondrial Regulator Sodium Dichloroacetate in Preclinical Studies In Vivo. Cancers. Aug 20 2019;11(8)doi:10.3390/cancers11081210
  349. Tataranni T, Agriesti F, Pacelli C, et al. Dichloroacetate Affects Mitochondrial Function and Stemness-Associated Properties in Pancreatic Cancer Cell Lines. Cells. May 18 2019;8(5)doi:10.3390/cells8050478
  350. Sun L, Moritake T, Ito K, et al. Metabolic analysis of radioresistant medulloblastoma stem-like clones and potential therapeutic targets. PLoS One. 2017;12(4):e0176162. doi:10.1371/journal.pone.0176162
  351. Khan A, Marier D, Marsden E, Andrews D, Eliaz I. A novel form of dichloroacetate therapy for patients with advanced cancer: a report of 3 cases. Alternative therapies in health and medicine. Oct 2014;20 Suppl 2:21-8.
  352. Strum SB, Adalsteinsson O, Black RR, Segal D, Peress NL, Waldenfels J. Case report: Sodium dichloroacetate (DCA) inhibition of the "Warburg Effect" in a human cancer patient: complete response in non-Hodgkin's lymphoma after disease progression with rituximab-CHOP. J Bioenerg Biomembr. Jun 2013;45(3):307-15. doi:10.1007/s10863-012-9496-2
  353. Tian DD, Bennett SK, Coupland LA, et al. GSTZ1 genotypes correlate with dichloroacetate pharmacokinetics and chronic side effects in multiple myeloma patients in a pilot phase 2 clinical trial. Pharmacology research & perspectives. Dec 2019;7(6):e00526. doi:10.1002/prp2.526
  354. James MO, Jahn SC, Zhong G, Smeltz MG, Hu Z, Stacpoole PW. Therapeutic applications of dichloroacetate and the role of glutathione transferase zeta-1. Pharmacology & therapeutics. Feb 2017;170:166-180. doi:10.1016/j.pharmthera.2016.10.018
  355. Younger J, Parkitny L, McLain D. The use of low-dose naltrexone (LDN) as a novel anti-inflammatory treatment for chronic pain. Clin Rheumatol . Apr 2014;33(4):451-9. doi:10.1007/s10067-014-2517-2
  356. Patten DK, Schultz BG, Berlau DJ. The Safety and Efficacy of Low-Dose Naltrexone in the Management of Chronic Pain and Inflammation in Multiple Sclerosis, Fibromyalgia, Crohn's Disease, and Other Chronic Pain Disorders. Pharmacotherapy. Mar 2018;38(3):382-389. doi:10.1002/phar.2086
  357. Toljan K, Vrooman B. Low-Dose Naltrexone (LDN)-Review of Therapeutic Utilization. Med Sci (Basel). Sep 21 2018;6(4)doi:10.3390/medsci6040082
  358. Li Z, You Y, Griffin N, Feng J, Shan F. Low-dose naltrexone (LDN): A promising treatment in immune-related diseases and cancer therapy. International immunopharmacology. Aug 2018;61:178-184. doi:10.1016/j.intimp.2018.05.020
  359. Boehncke S, Hardt K, Schadendorf D, Henschler R, Boehncke WH, Duthey B. Endogenous mu-opioid peptides modulate immune response towards malignant melanoma. Experimental dermatology. Jan 2011;20(1):24-8. doi:10.1111/j.1600-0625.2010.01158.x
  360. Lennon FE, Mirzapoiazova T, Mambetsariev B, Salgia R, Moss J, Singleton PA. Overexpression of the mu-opioid receptor in human non-small cell lung cancer promotes Akt and mTOR activation, tumor growth, and metastasis. Anesthesiology. Apr 2012;116(4):857-67. doi:10.1097/ALN.0b013e31824babe2
  361. Zylla D, Gourley BL, Vang D, et al. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer. Dec 1 2013;119(23):4103-10. doi:10.1002/cncr.28345
  362. Singleton PA, Mirzapoiazova T, Hasina R, Salgia R, Moss J. Increased mu-opioid receptor expression in metastatic lung cancer. British journal of anaesthesia. Jul 2014;113 Suppl 1:i103-8. doi:10.1093/bja/aeu165
  363. Yao YS, Yao RY, Zhuang LK, et al. MOR1 expression in gastric cancer: a biomarker associated with poor outcome. Clin Transl Sci. Apr 2015;8(2):137-42. doi:10.1111/cts.12246
  364. Qu N, Wang X, Meng Y, Shan F. Prospective oncotarget for gynecological cancer: Opioid growth factor (OGF) - opioid growth factor receptor (OGFr) axis. International immunopharmacology. Oct 2019;75:105723. doi:10.1016/j.intimp.2019.105723
  365. Donahue RN, McLaughlin PJ, Zagon IS. Low-dose naltrexone targets the opioid growth factor-opioid growth factor receptor pathway to inhibit cell proliferation: mechanistic evidence from a tissue culture model. Experimental biology and medicine (Maywood, NJ). Sep 2011;236(9):1036-50. doi:10.1258/ebm.2011.011121
  366. Wang R, Zhang Y, Shan F. Interaction of opioid growth factor (OGF) and opioid antagonist and their significance in cancer therapy. International immunopharmacology. Oct 2019;75:105785. doi:10.1016/j.intimp.2019.105785
  367. Selfridge BR, Wang X, Zhang Y, et al. Structure-Activity Relationships of (+)-Naltrexone-Inspired Toll-like Receptor 4 (TLR4) Antagonists. J Med Chem. Jun 25 2015;58(12):5038-52. doi:10.1021/acs.jmedchem.5b00426
  368. Wang X, Zhang Y, Peng Y, et al. Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4. Br J Pharmacol. Mar 2016;173(5):856-69. doi:10.1111/bph.13394
  369. Cant R, Dalgleish AG, Allen RL. Naltrexone Inhibits IL-6 and TNFalpha Production in Human Immune Cell Subsets following Stimulation with Ligands for Intracellular Toll-Like Receptors. Front Immunol. 2017;8:809. doi:10.3389/fimmu.2017.00809
  370. Ma M, Wang X, Liu N, Shan F, Feng Y. Low-dose naltrexone inhibits colorectal cancer progression and promotes apoptosis by increasing M1-type macrophages and activating the Bax/Bcl-2/caspase-3/PARP pathway. International immunopharmacology. Jun 2020;83:106388. doi:10.1016/j.intimp.2020.106388
  371. Liu WM, Scott KA, Dennis JL, Kaminska E, Levett AJ, Dalgleish AG. Naltrexone at low doses upregulates a unique gene expression not seen with normal doses: Implications for its use in cancer therapy. International journal of oncology. Aug 2016;49(2):793-802. doi:10.3892/ijo.2016.3567
  372. Aboalsoud A, El-Ghaiesh SH, Abd Elmonem FF, Salem ML, Abdel Rahman MN. The effect of low-dose naltrexone on solid Ehrlich carcinoma in mice: The role of OGFr, BCL2, and immune response. International immunopharmacology. Jan 2020;78:106068. doi:10.1016/j.intimp.2019.106068
  373. Donahue RN, McLaughlin PJ, Zagon IS. Low-dose naltrexone suppresses ovarian cancer and exhibits enhanced inhibition in combination with cisplatin. Experimental biology and medicine (Maywood, NJ). Jul 2011;236(7):883-95. doi:10.1258/ebm.2011.011096
  374. Machado MC, da Costa-Neto JM, Portela RD, et al. The effect of naltrexone as a carboplatin chemotherapy-associated drug on the immune response, quality of life and survival of dogs with mammary carcinoma. PLoS One. 2018;13(10):e0204830. doi:10.1371/journal.pone.0204830
  375. Berkson BM, Rubin DM, Berkson AJ. Revisiting the ALA/N (alpha-lipoic acid/low-dose naltrexone) protocol for people with metastatic and nonmetastatic pancreatic cancer: a report of 3 new cases. Integrative cancer therapies. Dec 2009;8(4):416-22. doi:10.1177/1534735409352082
  376. Berkson BM, Rubin DM, Berkson AJ. The long-term survival of a patient with pancreatic cancer with metastases to the liver after treatment with the intravenous alpha-lipoic acid/low-dose naltrexone protocol. Integrative cancer therapies. Mar 2006;5(1):83-9. doi:10.1177/1534735405285901
  377. Berkson BM, Calvo Riera F. The Long-Term Survival of a Patient With Stage IV Renal Cell Carcinoma Following an Integrative Treatment Approach Including the Intravenous alpha-Lipoic Acid/Low-Dose Naltrexone Protocol. Integrative cancer therapies. Sep 2018;17(3):986-993. doi:10.1177/1534735417747984
  378. Miskoff JA, Chaudhri M. Low Dose Naltrexone and Lung Cancer: A Case Report and Discussion. Cureus. Jul 5 2018;10(7):e2924. doi:10.7759/cureus.2924
  379. Rogosnitzky M, Finegold MJ, McLaughlin PJ, Zagon IS. Opioid growth factor (OGF) for hepatoblastoma: a novel non-toxic treatment. Investigational new drugs. Aug 2013;31(4):1066-70. doi:10.1007/s10637-012-9918-3
  380. Bolton M, Hodkinson A, Boda S, et al. Serious adverse events reported in placebo randomised controlled trials of oral naltrexone: a systematic review and meta-analysis. BMC Med. Jan 15 2019;17(1):10. doi:10.1186/s12916-018-1242-0
  381. Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov. Apr 2014;13(4):290-314. doi:10.1038/nrd4228
  382. Ahmad F, Murata T, Shimizu K, Degerman E, Maurice D, Manganiello V. Cyclic nucleotide phosphodiesterases: important signaling modulators and therapeutic targets. Oral diseases. Jan 2015;21(1):e25-50. doi:10.1111/odi.12275
  383. Armando RG, Mengual Gómez DL, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. International journal of oncology. Mar 2020;56(3):651-684. doi:10.3892/ijo.2020.4966
  384. Pantziarka P, Sukhatme V, Crispino S, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)-selective PDE5 inhibitors as anti-cancer agents. Ecancermedicalscience. 2018;12:824. doi:10.3332/ecancer.2018.824
  385. Houslay MD. Melanoma, Viagra, and PDE5 Inhibitors: Proliferation and Metastasis. Trends in cancer. Apr 2016;2(4):163-165. doi:10.1016/j.trecan.2016.02.007
  386. Christie A, Vera PL, Higgins M, Kumar S, Lane M, Preston D. Erectile Dysfunction Medications and Skin Cancer: An Analysis in US Veterans. Urology. Apr 2019;126:116-120. doi:10.1016/j.urology.2019.01.025
  387. Shkolyar E, Li S, Tang J, Eisenberg ML. Risk of Melanoma With Phosphodiesterase Type 5 Inhibitor Use Among Patients With Erectile Dysfunction, Pulmonary Hypertension, and Lower Urinary Tract Symptoms. The journal of sexual medicine. Jul 2018;15(7):982-989. doi:10.1016/j.jsxm.2018.05.002
  388. Wang JZ, Le S, Alexanian C, et al. No Causal Link between Phosphodiesterase Type 5 Inhibition and Melanoma. World J Mens Health. Sep 2019;37(3):313-321. doi:10.5534/wjmh.180050
  389. Feng S, Zhou L, Liu Q, et al. Are phosphodiesterase type 5 inhibitors associated with increased risk of melanoma?: A systematic review and meta-analysis. Medicine. Jan 2018;97(3):e9601. doi:10.1097/md.0000000000009601
  390. Sutton SS, Magagnoli J, Cummings TH, Hardin JW. The Association Between Phosphodiesterase-5 Inhibitors and Colorectal Cancer in a National Cohort of Patients. Clin Transl Gastroenterol. Jun 2020;11(6):e00173. doi:10.14309/ctg.0000000000000173
  391. Huang W, Sundquist J, Sundquist K, Ji J. Use of Phosphodiesterase 5 Inhibitors Is Associated With Lower Risk of Colorectal Cancer in Men With Benign Colorectal Neoplasms. Gastroenterology. Sep 2019;157(3):672-681.e4. doi:10.1053/j.gastro.2019.05.012
  392. Cea Soriano L, García Rodríguez LA. No association between use of phosphodiesterase 5 inhibitors and colorectal cancer in men with erectile dysfunction. Pharmacoepidemiology and drug safety. May 2020;29(5):605-608. doi:10.1002/pds.5000
  393. Huang W, Sundquist J, Sundquist K, Ji J. Phosphodiesterase-5 inhibitors use and risk for mortality and metastases among male patients with colorectal cancer. Nature communications. Jun 24 2020;11(1):3191. doi:10.1038/s41467-020-17028-4
  394. Jamnagerwalla J, Howard LE, Vidal AC, et al. The Association between Phosphodiesterase Type 5 Inhibitors and Prostate Cancer: Results from the REDUCE Study. J Urol. Sep 2016;196(3):715-20. doi:10.1016/j.juro.2016.03.172
  395. Wu Y, Qu X, Wang Y, et al. Effect of phosphodiesterase type 5 inhibitors on prostate cancer risk and biochemical recurrence after prostate cancer treatment: A systematic review and meta-analysis. Andrologia. Mar 2019;51(2):e13198. doi:10.1111/and.13198
  396. Aoun F, Slaoui A, Walid AHO, et al. Association between phosphodiesterase type 5 inhibitors and prostate cancer: A systematic review. Prog Urol. Oct 2018;28(12):560-566. doi:10.1016/j.purol.2018.07.004
  397. Michl U, Molfenter F, Graefen M, et al. Use of phosphodiesterase type 5 inhibitors may adversely impact biochemical recurrence after radical prostatectomy. J Urol. Feb 2015;193(2):479-83. doi:10.1016/j.juro.2014.08.111
  398. Gallina A, Bianchi M, Gandaglia G, et al. A Detailed Analysis of the Association Between Postoperative Phosphodiesterase Type 5 Inhibitor Use and the Risk of Biochemical Recurrence After Radical Prostatectomy. Eur Urol. Nov 2015;68(5):750-3. doi:10.1016/j.eururo.2015.02.002
  399. Loeb S, Folkvaljon Y, Robinson D, Schlomm T, Garmo H, Stattin P. Phosphodiesterase Type 5 Inhibitor Use and Disease Recurrence After Prostate Cancer Treatment. Eur Urol. Nov 2016;70(5):824-828. doi:10.1016/j.eururo.2015.12.013
  400. Chung JW, Kim JW, Ha YS, et al. The Effect of Phosphodiesterase 5 Inhibitor on Biochemical Recurrence Following Radical Prostatectomy in Patients with Prostate Cancer. Urol J. Jun 17 2019;16(3):255-259. doi:10.22037/uj.v16i3.4263
  401. He Q, Liao BH, Xiao KW, et al. Is there a relationship between phosphodiesterase type 5 inhibitors and biochemical recurrence after radical prostatectomy: a systematic review and meta-analysis. International urology and nephrology. Dec 2018;50(12):2113-2121. doi:10.1007/s11255-018-1982-y
  402. Hofer L, Radtke JP, Rapp C, et al. [Recurrence-free survival after radical prostatectomy and PDE-5 inhibitor intake]. Der Urologe Ausg A. Apr 2017;56(4):492-496. Rezidivfreies Überleben nach radikaler Prostatektomie und PDE-5-Hemmer-Einnahme. doi:10.1007/s00120-016-0267-2
  403. Treon SP, Tournilhac O, Branagan AR, et al. Clinical responses to sildenafil in Waldenstrom's macroglobulinemia. Clin Lymphoma. Dec 2004;5(3):205-7. doi:10.3816/clm.2004.n.029
  404. Huilgol NG, Jain A. A new indication of sildenafil in medicine: hypoxic cell sensitizer for penile cancer. Journal of cancer research and therapeutics. Jul-Sep 2006;2(3):132-5. doi:10.4103/0973-1482.27589
  405. Noonan KA, Ghosh N, Rudraraju L, Bui M, Borrello I. Targeting immune suppression with PDE5 inhibition in end-stage multiple myeloma. Cancer Immunol Res. Aug 2014;2(8):725-31. doi:10.1158/2326-6066.Cir-13-0213
  406. Pawelec G, Verschoor CP, Ostrand-Rosenberg S. Myeloid-Derived Suppressor Cells: Not Only in Tumor Immunity. Frontiers in immunology. 2019;10:1099-1099. doi:10.3389/fimmu.2019.01099
  407. Hassel JC, Jiang H, Bender C, et al. Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic Melanoma (TaMe). Oncoimmunology. 2017;6(9):e1326440. doi:10.1080/2162402x.2017.1326440
  408. Weed DT, Vella JL, Reis IM, et al. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. Jan 1 2015;21(1):39-48. doi:10.1158/1078-0432.Ccr-14-1711
  409. Califano JA, Khan Z, Noonan KA, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. Jan 1 2015;21(1):30-8. doi:10.1158/1078-0432.Ccr-14-1716
  410. ClinicalTrials.gov. Search: Cancer and Tadalafil. Accessed August 31, 2020. https://clinicaltrials.gov/ct2/results?term=tadalafil&cond=Cancer
  411. Verbaanderd C, Maes H, Schaaf MB, et al. Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience. 2017;11:781. doi:10.3332/ecancer.2017.781
  412. Cuomo F, Altucci L, Cobellis G. Autophagy Function and Dysfunction: Potential Drugs as Anti-Cancer Therapy. Cancers. Sep 29 2019;11(10)doi:10.3390/cancers11101465
  413. Cirone M, Gilardini Montani MS, Granato M, Garufi A, Faggioni A, D'Orazi G. Autophagy manipulation as a strategy for efficient anticancer therapies: possible consequences. Journal of experimental & clinical cancer research : CR. Jun 14 2019;38(1):262. doi:10.1186/s13046-019-1275-z
  414. Yun CW, Lee SH. The Roles of Autophagy in Cancer. International journal of molecular sciences. Nov 5 2018;19(11)doi:10.3390/ijms19113466
  415. Bishop E, Bradshaw TD. Autophagy modulation: a prudent approach in cancer treatment? Cancer chemotherapy and pharmacology. Dec 2018;82(6):913-922. doi:10.1007/s00280-018-3669-6
  416. Briceño E, Reyes S, Sotelo J. Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine. Neurosurgical focus. Feb 15 2003;14(2):e3. doi:10.3171/foc.2003.14.2.4
  417. Sotelo J, Briceño E, López-González MA. Adding chloroquine to conventional treatment for glioblastoma multiforme: a randomized, double-blind, placebo-controlled trial. Ann Intern Med. Mar 7 2006;144(5):337-43. doi:10.7326/0003-4819-144-5-200603070-00008
  418. Briceño E, Calderon A, Sotelo J. Institutional experience with chloroquine as an adjuvant to the therapy for glioblastoma multiforme. Surgical neurology. Apr 2007;67(4):388-91. doi:10.1016/j.surneu.2006.08.080
  419. Levy JM, Thompson JC, Griesinger AM, et al. Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors. Cancer Discov. Jul 2014;4(7):773-80. doi:10.1158/2159-8290.Cd-14-0049
  420. Mulcahy Levy JM, Zahedi S, Griesinger AM, et al. Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors. Elife. Jan 17 2017;6doi:10.7554/eLife.19671
  421. Rojas-Puentes LL, Gonzalez-Pinedo M, Crismatt A, et al. Phase II randomized, double-blind, placebo-controlled study of whole-brain irradiation with concomitant chloroquine for brain metastases. Radiation oncology (London, England). Sep 8 2013;8:209. doi:10.1186/1748-717x-8-209
  422. Eldredge HB, DeNittis A, DuHadaway JB, Chernick M, Metz R, Prendergast GC. Concurrent whole brain radiotherapy and short-course chloroquine in patients with brain metastases: a pilot trial. Journal of Radiation Oncology. 2013/09/01 2013;2(3):315-321. doi:10.1007/s13566-013-0111-x
  423. Montanari F, Lu M, Marcus S, Saran A, Malankar A, Mazumder A. A Phase II Trial of Chloroquine in Combination with Bortezomib and Cyclophosphamide in Patients with Relapsed and Refractory Multiple Myeloma. Blood. 2014;124(21):5775-5775. doi:10.1182/blood.V124.21.5775.5775
  424. Kyle RA, Seligman BR, Wallace HJ, Jr., Silver RT, Glidewell O, Holland JF. Mutiple myeloma resistant to melphalan (NSC-8806) treated with cyclophosphamide (NSC-26271), prednisone (NSC-10023), and chloroquine (NSC-187208). Cancer Chemother Rep. May-Jun 1975;59(3):557-62.
  425. Samaras P, Tusup M, Nguyen-Kim TDL, et al. Phase I study of a chloroquine-gemcitabine combination in patients with metastatic or unresectable pancreatic cancer. Cancer chemotherapy and pharmacology. Nov 2017;80(5):1005-1012. doi:10.1007/s00280-017-3446-y
  426. Chi KH, Ko HL, Yang KL, Lee CY, Chi MS, Kao SJ. Addition of rapamycin and hydroxychloroquine to metronomic chemotherapy as a second line treatment results in high salvage rates for refractory metastatic solid tumors: a pilot safety and effectiveness analysis in a small patient cohort. Oncotarget. Jun 30 2015;6(18):16735-45. doi:10.18632/oncotarget.3793
  427. Mahalingam D, Mita M, Sarantopoulos J, et al. Combined autophagy and HDAC inhibition: a phase I safety, tolerability, pharmacokinetic, and pharmacodynamic analysis of hydroxychloroquine in combination with the HDAC inhibitor vorinostat in patients with advanced solid tumors. Autophagy. Aug 2014;10(8):1403-14. doi:10.4161/auto.29231
  428. Mehnert JM, Kaveney AD, Malhotra J, et al. A phase I trial of MK-2206 and hydroxychloroquine in patients with advanced solid tumors. Cancer chemotherapy and pharmacology. Oct 2019;84(4):899-907. doi:10.1007/s00280-019-03919-x
  429. Wolpin BM, Rubinson DA, Wang X, et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. The oncologist. Jun 2014;19(6):637-8. doi:10.1634/theoncologist.2014-0086
  430. Boone BA, Bahary N, Zureikat AH, et al. Safety and Biologic Response of Pre-operative Autophagy Inhibition in Combination with Gemcitabine in Patients with Pancreatic Adenocarcinoma. Annals of surgical oncology. Dec 2015;22(13):4402-10. doi:10.1245/s10434-015-4566-4
  431. Zeh HJ, Bahary N, Boone BA, et al. A Randomized Phase II Preoperative Study of Autophagy Inhibition with High-Dose Hydroxychloroquine and Gemcitabine/Nab-Paclitaxel in Pancreatic Cancer Patients. Clin Cancer Res. Jul 1 2020;26(13):3126-3134. doi:10.1158/1078-0432.Ccr-19-4042
  432. Karasic TB, O'Hara MH, Loaiza-Bonilla A, et al. Effect of Gemcitabine and nab-Paclitaxel With or Without Hydroxychloroquine on Patients With Advanced Pancreatic Cancer: A Phase 2 Randomized Clinical Trial. JAMA oncology. Jul 1 2019;5(7):993-998. doi:10.1001/jamaoncol.2019.0684
  433. Patel S, Hurez V, Nawrocki ST, et al. Vorinostat and hydroxychloroquine improve immunity and inhibit autophagy in metastatic colorectal cancer. Oncotarget. Sep 13 2016;7(37):59087-59097. doi:10.18632/oncotarget.10824
  434. Vogl DT, Stadtmauer EA, Tan KS, et al. Combined autophagy and proteasome inhibition: a phase 1 trial of hydroxychloroquine and bortezomib in patients with relapsed/refractory myeloma. Autophagy. Aug 2014;10(8):1380-90. doi:10.4161/auto.29264
  435. Chi MS, Lee CY, Huang SC, et al. Double autophagy modulators reduce 2-deoxyglucose uptake in sarcoma patients. Oncotarget. Oct 6 2015;6(30):29808-17. doi:10.18632/oncotarget.5060
  436. Rangwala R, Leone R, Chang YC, et al. Phase I trial of hydroxychloroquine with dose-intense temozolomide in patients with advanced solid tumors and melanoma. Autophagy. Aug 2014;10(8):1369-79. doi:10.4161/auto.29118
  437. Rangwala R, Chang YC, Hu J, et al. Combined MTOR and autophagy inhibition: phase I trial of hydroxychloroquine and temsirolimus in patients with advanced solid tumors and melanoma. Autophagy. Aug 2014;10(8):1391-402. doi:10.4161/auto.29119
  438. Goldberg SB, Supko JG, Neal JW, et al. A phase I study of erlotinib and hydroxychloroquine in advanced non-small-cell lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer . Oct 2012;7(10):1602-8. doi:10.1097/JTO.0b013e318262de4a
  439. Malhotra J, Jabbour S, Orlick M, et al. Phase Ib/II study of hydroxychloroquine in combination with chemotherapy in patients with metastatic non-small cell lung cancer (NSCLC). Cancer Treat Res Commun. 2019;21:100158. doi:10.1016/j.ctarc.2019.100158
  440. Haas NB, Appleman LJ, Stein M, et al. Autophagy Inhibition to Augment mTOR Inhibition: a Phase I/II Trial of Everolimus and Hydroxychloroquine in Patients with Previously Treated Renal Cell Carcinoma. Clin Cancer Res. Apr 1 2019;25(7):2080-2087. doi:10.1158/1078-0432.Ccr-18-2204
  441. Xu R, Ji Z, Xu C, Zhu J. The clinical value of using chloroquine or hydroxychloroquine as autophagy inhibitors in the treatment of cancers: A systematic review and meta-analysis. Medicine. Nov 2018;97(46):e12912. doi:10.1097/md.0000000000012912
  442. Stokkermans TJ, Goyal A, Bansal P, Trichonas G. Chloroquine And Hydroxychloroquine Toxicity. StatPearls. StatPearls Publishing Copyright © 2020, StatPearls Publishing LLC.; 2020.
  443. Sapp JL, Alqarawi W, MacIntyre CJ, et al. Guidance on Minimizing Risk of Drug-Induced Ventricular Arrhythmia During Treatment of COVID-19: A Statement from the Canadian Heart Rhythm Society. The Canadian journal of cardiology. Jun 2020;36(6):948-951. doi:10.1016/j.cjca.2020.04.003
  444. Guevara B, Cogdill AG. Helicobacter pylori: A Review of Current Diagnostic and Management Strategies. Dig Dis Sci. Jul 2020;65(7):1917-1931. doi:10.1007/s10620-020-06193-7
  445. Laird-Fick HS, Saini S, Hillard JR. Gastric adenocarcinoma: the role of Helicobacter pylori in pathogenesis and prevention efforts. Postgrad Med J. Aug 2016;92(1090):471-7. doi:10.1136/postgradmedj-2016-133997
  446. Van Nuffel AM, Sukhatme V, Pantziarka P, Meheus L, Sukhatme VP, Bouche G. Repurposing Drugs in Oncology (ReDO)-clarithromycin as an anti-cancer agent. Ecancermedicalscience. 2015;9:513. doi:10.3332/ecancer.2015.513
  447. Petroni G, Stefanini M, Pillozzi S, Crociani O, Becchetti A, Arcangeli A. Data describing the effects of the Macrolide Antibiotic Clarithromycin on preclinical mouse models of Colorectal Cancer. Data Brief. Oct 2019;26:104406. doi:10.1016/j.dib.2019.104406
  448. Hirasawa K, Moriya S, Miyahara K, et al. Macrolide Antibiotics Exhibit Cytotoxic Effect under Amino Acid-Depleted Culture Condition by Blocking Autophagy Flux in Head and Neck Squamous Cell Carcinoma Cell Lines. PLoS One. 2016;11(12):e0164529. doi:10.1371/journal.pone.0164529
  449. Zhou L, Lin S, Ding S, et al. Relationship of Helicobacter pylori eradication with gastric cancer and gastric mucosal histological changes: a 10-year follow-up study. Chin Med J (Engl). 2014;127(8):1454-8.
  450. Choi JM, Kim SG, Choi J, et al. Effects of Helicobacter pylori eradication for metachronous gastric cancer prevention: a randomized controlled trial. Gastrointest Endosc. Sep 2018;88(3):475-485.e2. doi:10.1016/j.gie.2018.05.009
  451. Choi IJ, Kook MC, Kim YI, et al. Helicobacter pylori Therapy for the Prevention of Metachronous Gastric Cancer. The New England journal of medicine. Mar 22 2018;378(12):1085-1095. doi:10.1056/NEJMoa1708423
  452. Graham DY, Dore MP. Helicobacter pylori therapy: a paradigm shift. Expert review of anti-infective therapy. Jun 2016;14(6):577-85. doi:10.1080/14787210.2016.1178065
  453. Puig N, Hernández MT, Rosinol Dachs L, et al. Randomized Trial of Lenalidomide and Dexamethasone Versus Clarythromycin, Lenalidomide and Dexamethasone As First Line Treatment in Patients with Multiple Myeloma Not Candidates for Autologous Stem Cell Transplantation: Results of the GEM-Claridex Clinical Trial. Blood. 2019;134(Supplement_1):694-694. doi:10.1182/blood-2019-123997
  454. Holmberg LA, Becker PS, Bensinger W. Results from Two Consecutive Studies of Consolidation Therapy after Autologous Transplant for Multiple Myeloma: Thalidomide, Dexamethasone, and Clarithromycin or Lenalidomide, Dexamethasone, and Clarithromycin. Acta haematologica. 2017;137(3):123-131. doi:10.1159/000455937
  455. Gay F, Rajkumar SV, Coleman M, et al. Clarithromycin (Biaxin)-lenalidomide-low-dose dexamethasone (BiRd) versus lenalidomide-low-dose dexamethasone (Rd) for newly diagnosed myeloma. Am J Hematol. Sep 2010;85(9):664-9. doi:10.1002/ajh.21777
  456. Rossi A, Mark T, Jayabalan D, et al. BiRd (clarithromycin, lenalidomide, dexamethasone): an update on long-term lenalidomide therapy in previously untreated patients with multiple myeloma. Blood. Mar 14 2013;121(11):1982-5. doi:10.1182/blood-2012-08-448563
  457. Coleman M, Leonard J, Lyons L, et al. BLT-D (clarithromycin [Biaxin], low-dose thalidomide, and dexamethasone) for the treatment of myeloma and Waldenström's macroglobulinemia. Leuk Lymphoma. Sep 2002;43(9):1777-82. doi:10.1080/1042819021000006303
  458. Mark TM, Bowman IA, Rossi AC, et al. Thalidomide, clarithromycin, lenalidomide and dexamethasone therapy in newly diagnosed, symptomatic multiple myeloma. Leuk Lymphoma. Dec 2014;55(12):2842-9. doi:10.3109/10428194.2014.896005
  459. Shaulov A, Ganzel C, Benyamini N, et al. Progressive refractory light chain amyloidosis and multiple myeloma patients are responsive to the addition of clarithromycin to IMiD based therapy. Am J Hematol. Feb 2017;92(2):131-135. doi:10.1002/ajh.24596
  460. Mark TM, Forsberg PA, Rossi AC, et al. Phase 2 study of clarithromycin, pomalidomide, and dexamethasone in relapsed or refractory multiple myeloma. Blood Adv. Feb 26 2019;3(4):603-611. doi:10.1182/bloodadvances.2018028027
  461. Nielsen LK, Klausen TW, Jarden M, et al. Clarithromycin added to bortezomib-cyclophosphamide-dexamethasone impairs health-related quality of life in multiple myeloma patients. Eur J Haematol. Jan 2019;102(1):70-78. doi:10.1111/ejh.13175
  462. Gregersen H, Do T, Kristensen IB, et al. A randomized placebo-controlled phase II study of clarithromycin or placebo combined with VCD induction therapy prior to high-dose melphalan with stem cell support in patients with newly diagnosed multiple myeloma. Exp Hematol Oncol. 2018;7:18. doi:10.1186/s40164-018-0110-0
  463. Ohe M, Hashino S, Shida H, Horita T, Sugiura M. A case of Waldenström's macroglobulinemia treated using clarithromycin and prednisolone. Transl Clin Pharmacol. Sep 2017;25(3):134-137. doi:10.12793/tcp.2017.25.3.134
  464. Dimopoulos MA, Tsatalas C, Zomas A, et al. Treatment of Waldenstrom's macroglobulinemia with single-agent thalidomide or with the combination of clarithromycin, thalidomide and dexamethasone. Seminars in oncology. Apr 2003;30(2):265-9. doi:10.1053/sonc.2003.50079
  465. Coleman M, Leonard J, Lyons L, Szelenyi H, Niesvizky R. Treatment of Waldenstrom's macroglobulinemia with clarithromycin, low-dose thalidomide, and dexamethasone. Seminars in oncology. Apr 2003;30(2):270-4. doi:10.1053/sonc.2003.50044
  466. Guo Q, Guo S, Zhang Y. Treatment of gastric MALT lymphoma with a focus on Helicobacter pylori eradication. Int J Hematol. Jun 2013;97(6):735-42. doi:10.1007/s12185-013-1348-2
  467. Fischbach W, Goebeler-Kolve ME, Dragosics B, Greiner A, Stolte M. Long term outcome of patients with gastric marginal zone B cell lymphoma of mucosa associated lymphoid tissue (MALT) following exclusive Helicobacter pylori eradication therapy: experience from a large prospective series. Gut. Jan 2004;53(1):34-7. doi:10.1136/gut.53.1.34
  468. Ruskoné-Fourmestraux A, Lavergne A, Aegerter PH, et al. Predictive factors for regression of gastric MALT lymphoma after anti-Helicobacter pylori treatment. Gut. Mar 2001;48(3):297-303. doi:10.1136/gut.48.3.297
  469. Cavanna L, Pagani R, Seghini P, Zangrandi A, Paties C. High grade B-cell gastric lymphoma with complete pathologic remission after eradication of Helicobacter pylori infection: report of a case and review of the literature. World journal of surgical oncology. Mar 19 2008;6:35. doi:10.1186/1477-7819-6-35
  470. Chen LT, Lin JT, Shyu RY, et al. Prospective study of Helicobacter pylori eradication therapy in stage I(E) high-grade mucosa-associated lymphoid tissue lymphoma of the stomach. J Clin Oncol. Nov 15 2001;19(22):4245-51. doi:10.1200/jco.2001.19.22.4245
  471. Ferreri AJ, Sassone M, Kiesewetter B, et al. High-dose clarithromycin is an active monotherapy for patients with relapsed/refractory extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue (MALT): the HD-K phase II trial. Ann Oncol. Aug 2015;26(8):1760-5. doi:10.1093/annonc/mdv214
  472. Saad AS, Shaheen SM, Elhamamsy MH, Badary OA. An open-label randomized controlled phase II study of clarithromycin (CL) plus CVP in patients (pts) with previously untreated stage III/IV indolent non Hodgkin lymphoma (NHL). Journal of Clinical Oncology. 2014;32(15_suppl):e19510-e19510. doi:10.1200/jco.2014.32.15_suppl.e19510
  473. Mikasa K, Sawaki M, Kita E, et al. Significant survival benefit to patients with advanced non-small-cell lung cancer from treatment with clarithromycin. Chemotherapy. Jul-Aug 1997;43(4):288-96. doi:10.1159/000239580
  474. Sakamoto M, Mikasa K, Majima T, et al. Anti-cachectic effect of clarithromycin for patients with unresectable non-small cell lung cancer. Chemotherapy. Dec 2001;47(6):444-51. doi:10.1159/000048556
  475. Sakamoto M, Mikasa K, Majima T, et al. [Usefulness of clarithromycin in paitents with unresectable non-small-cell lung cancer--clinical evaluation of those who survived for a long time and those succumbed in a short period]. Jpn J Antibiot. Jan 2000;53 Suppl A:56-9.
  476. Majima T, Mikasa K, Sakamoto M, et al. [Administration of clarithromycin (CAM) to non-small cell lung cancer]. Jpn J Antibiot. Jan 2000;53 Suppl A:52-5.
  477. Sakamoto M, Mikasa K, Sawaki M, et al. [Efficacy of clarithromycin (CAM) in patients with inoperable non-small-cell lung cancer]. Jpn J Antibiot. Jan 1998;51 Suppl A:50-2.
  478. Sakamoto M, Mikasa K, Hamada K, et al. [Effect of clarithromycin treatment of natural killer cell activity in patients with advanced non-small cell lung cancer]. Gan to kagaku ryoho Cancer & chemotherapy. Dec 1998;25(14):2259-66.
  479. Ieshiro R, Goto G, Ota T, Yuasa K, Iguchi M, Okamura J. [Effect of clarithromycin in extending the life expectancy of patients with non-small-cell lung cancer--the second report]. Jpn J Antibiot. Jan 1998;51 Suppl A:47-9.
  480. Sakamoto M, Mikasa K, Hamada K, et al. [Efficacy of clarithromycin on cancer cachexia in patients with primary non-small cell lung cancer]. Jpn J Antibiot. Mar 1997;50 Suppl A:38-41.
  481. Ieshiro R, Goto G, Ota T, Yuasa K, Iguchi M, Okamura J. [Effect of clarithromycin in extending the life expectancy of patients with non-small-cell lung cancer]. Jpn J Antibiot. Mar 1997;50 Suppl A:49-52.
  482. Tsuji D, Kamezato M, Daimon T, et al. Retrospective analysis of severe neutropenia in patients receiving concomitant administration of docetaxel and clarithromycin. Chemotherapy. 2013;59(6):407-13. doi:10.1159/000362437
  483. Yano R, Tani D, Watanabe K, et al. Evaluation of potential interaction between vinorelbine and clarithromycin. Ann Pharmacother. Mar 2009;43(3):453-8. doi:10.1345/aph.1L432
  484. Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-itraconazole as an anti-cancer agent. Ecancermedicalscience. 2015;9:521. doi:10.3332/ecancer.2015.521
  485. Li K, Fang D, Xiong Z, Luo R. Inhibition of the hedgehog pathway for the treatment of cancer using Itraconazole. OncoTargets and therapy. 2019;12:6875-6886. doi:10.2147/ott.S223119
  486. Skoda AM, Simovic D, Karin V, Kardum V, Vranic S, Serman L. The role of the Hedgehog signaling pathway in cancer: A comprehensive review. Bosnian journal of basic medical sciences. Feb 20 2018;18(1):8-20. doi:10.17305/bjbms.2018.2756
  487. Cortes JE, Gutzmer R, Kieran MW, Solomon JA. Hedgehog signaling inhibitors in solid and hematological cancers. Cancer treatment reviews. Jun 2019;76:41-50. doi:10.1016/j.ctrv.2019.04.005
  488. Liu R, Li J, Zhang T, et al. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: involvement of abnormal cholesterol trafficking. Autophagy. Jul 2014;10(7):1241-55. doi:10.4161/auto.28912
  489. Bae SH, Park JH, Choi HG, Kim H, Kim SH. Imidazole Antifungal Drugs Inhibit the Cell Proliferation and Invasion of Human Breast Cancer Cells. Biomolecules & therapeutics. Sep 1 2018;26(5):494-502. doi:10.4062/biomolther.2018.042
  490. Wang X, Wei S, Zhao Y, et al. Anti-proliferation of breast cancer cells with itraconazole: Hedgehog pathway inhibition induces apoptosis and autophagic cell death. Cancer letters. Jan 28 2017;385:128-136. doi:10.1016/j.canlet.2016.10.034
  491. Ueda T, Tsubamoto H, Inoue K, Sakata K, Shibahara H, Sonoda T. Itraconazole Modulates Hedgehog, WNT/β-catenin, as well as Akt Signalling, and Inhibits Proliferation of Cervical Cancer Cells. Anticancer research. Jul 2017;37(7):3521-3526. doi:10.21873/anticanres.11720
  492. Tsubamoto H, Inoue K, Sakata K, et al. Itraconazole Inhibits AKT/mTOR Signaling and Proliferation in Endometrial Cancer Cells. Anticancer research. Feb 2017;37(2):515-519. doi:10.21873/anticanres.11343
  493. Inoue K, Tsubamoto H, Sakata K, et al. Expression of Hedgehog Signals and Growth Inhibition by Itraconazole in Endometrial Cancer. Anticancer research. Jan 2016;36(1):149-53.
  494. Choi CH, Ryu JY, Cho YJ, et al. The anti-cancer effects of itraconazole in epithelial ovarian cancer. Sci Rep. Jul 26 2017;7(1):6552. doi:10.1038/s41598-017-06510-7
  495. Ban L, Mei T, Su Q, et al. Anti-fungal drug itraconazole exerts anti-cancer effects in oral squamous cell carcinoma via suppressing Hedgehog pathway. Life Sci. Aug 1 2020;254:117695. doi:10.1016/j.lfs.2020.117695
  496. Chen MB, Liu YY, Xing ZY, et al. Itraconazole-Induced Inhibition on Human Esophageal Cancer Cell Growth Requires AMPK Activation. Molecular cancer therapeutics. Jun 2018;17(6):1229-1239. doi:10.1158/1535-7163.Mct-17-1094
  497. Chen K, Cheng L, Qian W, et al. Itraconazole inhibits invasion and migration of pancreatic cancer cells by suppressing TGF-β/SMAD2/3 signaling. Oncology reports. Apr 2018;39(4):1573-1582. doi:10.3892/or.2018.6281
  498. Hu Q, Hou YC, Huang J, Fang JY, Xiong H. Itraconazole induces apoptosis and cell cycle arrest via inhibiting Hedgehog signaling in gastric cancer cells. Journal of experimental & clinical cancer research : CR . Apr 11 2017;36(1):50. doi:10.1186/s13046-017-0526-0
  499. Deng H, Huang L, Liao Z, Liu M, Li Q, Xu R. Itraconazole inhibits the Hedgehog signaling pathway thereby inducing autophagy-mediated apoptosis of colon cancer cells. Cell Death Dis. Jul 17 2020;11(7):539. doi:10.1038/s41419-020-02742-0
  500. Liang G, Liu M, Wang Q, et al. Itraconazole exerts its anti-melanoma effect by suppressing Hedgehog, Wnt, and PI3K/mTOR signaling pathways. Oncotarget. Apr 25 2017;8(17):28510-28525. doi:10.18632/oncotarget.15324
  501. Kelly RJ, Ansari AM, Miyashita T, et al. Targeting the Hedgehog Pathway Using Itraconazole to Prevent Progression of Barrett's Esophagus to Invasive Esophageal Adenocarcinoma. Ann Surg. Jul 8 2019;doi:10.1097/sla.0000000000003455
  502. Lan K, Yan R, Zhu K, et al. Itraconazole inhibits the proliferation of gastric cancer cells in vitro and improves patient survival. Oncology letters. Sep 2018;16(3):3651-3657. doi:10.3892/ol.2018.9072
  503. Wang J, Xu X, Zhou R, Guo K. [Effects of itraconazole plus doxorubicin on proliferation and apoptosis in acute myeloid leukemia cells]. Zhonghua yi xue za zhi. Jan 27 2015;95(4):299-305.
  504. Vreugdenhil G, Raemaekers JMM, van Dijke BJ, de Pauw BE. Itraconazole and multidrug resistance: Possible effects on remission rate and disease-free survival in acute leukemia. Annals of hematology. 1993/09/01 1993;67(3):107-109. doi:10.1007/BF01701730
  505. Tsubamoto H, Sonoda T, Ikuta S, Tani S, Inoue K, Yamanaka N. Combination Chemotherapy with Itraconazole for Treating Metastatic Pancreatic Cancer in the Second-line or Additional Setting. Anticancer research. Jul 2015;35(7):4191-6.
  506. Tsubamoto H, Sonoda T, Ikuta S, Tani S, Inoue K, Yamanaka N. Impact of Itraconazole After First-line Chemotherapy on Survival of Patients with Metastatic Biliary Tract Cancer. Anticancer research. Sep 2015;35(9):4923-7.
  507. Tsubamoto H, Sonoda T, Inoue K. Impact of itraconazole on the survival of heavily pre-treated patients with triple-negative breast cancer. Anticancer research. Jul 2014;34(7):3839-44.
  508. Tsubamoto H, Sonoda T, Yamasaki M, Inoue K. Impact of combination chemotherapy with itraconazole on survival for patients with recurrent or persistent ovarian clear cell carcinoma. Anticancer research. Apr 2014;34(4):2007-14.
  509. Tsubamoto H, Sonoda T, Yamasaki M, Inoue K. Impact of combination chemotherapy with itraconazole on survival of patients with refractory ovarian cancer. Anticancer research. May 2014;34(5):2481-7.
  510. Tsubamoto H, Ueda T, Inoue K, Sakata K, Shibahara H, Sonoda T. Repurposing itraconazole as an anticancer agent. Oncology letters. Aug 2017;14(2):1240-1246. doi:10.3892/ol.2017.6325
  511. Kim DJ, Kim J, Spaunhurst K, et al. Open-label, exploratory phase II trial of oral itraconazole for the treatment of basal cell carcinoma. J Clin Oncol. Mar 10 2014;32(8):745-51. doi:10.1200/jco.2013.49.9525
  512. Ally MS, Ransohoff K, Sarin K, et al. Effects of Combined Treatment With Arsenic Trioxide and Itraconazole in Patients With Refractory Metastatic Basal Cell Carcinoma. JAMA Dermatol. Apr 2016;152(4):452-6. doi:10.1001/jamadermatol.2015.5473
  513. Yoon J, Apicelli AJ, 3rd, Pavlopoulos TV. Intracranial regression of an advanced basal cell carcinoma using sonidegib and itraconazole after failure with vismodegib. JAAD Case Rep. Jan 2018;4(1):10-12. doi:10.1016/j.jdcr.2017.11.001
  514. Antonarakis ES, Heath EI, Smith DC, et al. Repurposing itraconazole as a treatment for advanced prostate cancer: a noncomparative randomized phase II trial in men with metastatic castration-resistant prostate cancer. The oncologist. 2013;18(2):163-73. doi:10.1634/theoncologist.2012-314
  515. Lee M, Hong H, Kim W, et al. Itraconazole as a Noncastrating Treatment for Biochemically Recurrent Prostate Cancer: A Phase 2 Study. Clinical genitourinary cancer. Feb 2019;17(1):e92-e96. doi:10.1016/j.clgc.2018.09.013
  516. Suzman DL, Antonarakis ES. High-dose itraconazole as a noncastrating therapy for a patient with biochemically recurrent prostate cancer. Clinical genitourinary cancer. Apr 2014;12(2):e51-3. doi:10.1016/j.clgc.2013.11.015
  517. Sawasaki M, Tsubamoto H, Nakamoto Y, Kakuno A, Sonoda T. S-1, Oxaliplatin, Nab-paclitaxel and Itraconazole for Conversion Surgery for Advanced or Recurrent Gastric Cancer. Anticancer research. Feb 2020;40(2):991-997. doi:10.21873/anticanres.14033
  518. Lockhart NR, Waddell JA, Schrock NE. Itraconazole therapy in a pancreatic adenocarcinoma patient: A case report. J Oncol Pharm Pract. Jun 2016;22(3):528-32. doi:10.1177/1078155215572931
  519. Rudin CM, Brahmer JR, Juergens RA, et al. Phase 2 study of pemetrexed and itraconazole as second-line therapy for metastatic nonsquamous non-small-cell lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer . May 2013;8(5):619-23. doi:10.1097/JTO.0b013e31828c3950
  520. Gerber DE, Putnam WC, Fattah FJ, et al. Concentration-dependent early anti-vascular and anti-tumor effects of itraconazole in non-small cell lung cancer. Clin Cancer Res. Aug 26 2020;doi:10.1158/1078-0432.Ccr-20-1916
  521. Benitez LL, Carver PL. Adverse Effects Associated with Long-Term Administration of Azole Antifungal Agents. Drugs. Jun 2019;79(8):833-853. doi:10.1007/s40265-019-01127-8
  522. Li Y, Pasunooti KK, Li RJ, et al. Novel Tetrazole-Containing Analogues of Itraconazole as Potent Antiangiogenic Agents with Reduced Cytochrome P450 3A4 Inhibition. J Med Chem. Dec 27 2018;61(24):11158-11168. doi:10.1021/acs.jmedchem.8b01252
  523. Guerini AE, Triggiani L, Maddalo M, et al. Mebendazole as a Candidate for Drug Repurposing in Oncology: An Extensive Review of Current Literature. Cancers. Aug 31 2019;11(9)doi:10.3390/cancers11091284
  524. Arnst KE, Banerjee S, Chen H, et al. Current advances of tubulin inhibitors as dual acting small molecules for cancer therapy. Medicinal research reviews. Jul 2019;39(4):1398-1426. doi:10.1002/med.21568
  525. Larsen AR, Bai RY, Chung JH, et al. Repurposing the antihelmintic mebendazole as a hedgehog inhibitor. Molecular cancer therapeutics . Jan 2015;14(1):3-13. doi:10.1158/1535-7163.Mct-14-0755-t
  526. Sung SJ, Kim HK, Hong YK, Joe YA. Autophagy Is a Potential Target for Enhancing the Anti-Angiogenic Effect of Mebendazole in Endothelial Cells. Biomolecules & therapeutics. Jan 1 2019;27(1):117-125. doi:10.4062/biomolther.2018.222
  527. Blom K, Senkowski W, Jarvius M, et al. The anticancer effect of mebendazole may be due to M1 monocyte/macrophage activation via ERK1/2 and TLR8-dependent inflammasome activation. Immunopharmacology and immunotoxicology. Aug 2017;39(4):199-210. doi:10.1080/08923973.2017.1320671
  528. Nath J, Paul R, Ghosh SK, Paul J, Singha B, Debnath N. Drug repurposing and relabeling for cancer therapy: Emerging benzimidazole antihelminthics with potent anticancer effects. Life Sci. Aug 8 2020;258:118189. doi:10.1016/j.lfs.2020.118189
  529. De Witt M, Gamble A, Hanson D, et al. Repurposing Mebendazole as a Replacement for Vincristine for the Treatment of Brain Tumors. Molecular medicine (Cambridge, Mass). Apr 2017;23:50-56. doi:10.2119/molmed.2017.00011
  530. Lai SR, Castello SA, Robinson AC, Koehler JW. In vitro anti-tubulin effects of mebendazole and fenbendazole on canine glioma cells. Vet Comp Oncol. Dec 2017;15(4):1445-1454. doi:10.1111/vco.12288
  531. Bai RY, Staedtke V, Rudin CM, Bunz F, Riggins GJ. Effective treatment of diverse medulloblastoma models with mebendazole and its impact on tumor angiogenesis. Neuro Oncol. Apr 2015;17(4):545-54. doi:10.1093/neuonc/nou234
  532. Bai RY, Staedtke V, Aprhys CM, Gallia GL, Riggins GJ. Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme. Neuro Oncol. Sep 2011;13(9):974-82. doi:10.1093/neuonc/nor077
  533. Kipper FC, Silva AO, Marc AL, et al. Vinblastine and antihelmintic mebendazole potentiate temozolomide in resistant gliomas. Investigational new drugs. Apr 2018;36(2):323-331. doi:10.1007/s10637-017-0503-7
  534. Zhang L, Bochkur Dratver M, Yazal T, et al. Mebendazole Potentiates Radiation Therapy in Triple-Negative Breast Cancer. International journal of radiation oncology, biology, physics. Jan 1 2019;103(1):195-207. doi:10.1016/j.ijrobp.2018.08.046
  535. Celestino Pinto L, de Fátima Aquino Moreira-Nunes C, Soares BM, Burbano RMR, de Lemos JAR, Montenegro RC. Mebendazole, an antiparasitic drug, inhibits drug transporters expression in preclinical model of gastric peritoneal carcinomatosis. Toxicol In Vitro. Sep 2017;43:87-91. doi:10.1016/j.tiv.2017.06.007
  536. Pinto LC, Soares BM, Pinheiro Jde J, et al. The anthelmintic drug mebendazole inhibits growth, migration and invasion in gastric cancer cell model. Toxicol In Vitro. Dec 2015;29(8):2038-44. doi:10.1016/j.tiv.2015.08.007
  537. Williamson T, Bai RY, Staedtke V, Huso D, Riggins GJ. Mebendazole and a non-steroidal anti-inflammatory combine to reduce tumor initiation in a colon cancer preclinical model. Oncotarget. Oct 18 2016;7(42):68571-68584. doi:10.18632/oncotarget.11851
  538. Nygren P, Fryknäs M, Agerup B, Larsson R. Repositioning of the anthelmintic drug mebendazole for the treatment for colon cancer. Journal of cancer research and clinical oncology. Dec 2013;139(12):2133-40. doi:10.1007/s00432-013-1539-5
  539. Rushworth LK, Hewit K, Munnings-Tomes S, et al. Repurposing screen identifies mebendazole as a clinical candidate to synergise with docetaxel for prostate cancer treatment. British journal of cancer. Feb 2020;122(4):517-527. doi:10.1038/s41416-019-0681-5
  540. Coyne CP, Jones T, Bear R. Anti-Neoplastic Cytotoxicity of Gemcitabine-(C(4)-amide)-[anti-EGFR] in Dual-combination with Epirubicin-(C(3)-amide)-[anti-HER2/neu] against Chemotherapeutic-Resistant Mammary Adenocarcinoma (SKBr-3) and the Complementary Effect of Mebendazole. J Cancer Res Ther Oncol. Apr 9 2014;2(1)doi:10.17303/jcrto.2014.203
  541. Younis NS, Ghanim AMH, Saber S. Mebendazole augments sensitivity to sorafenib by targeting MAPK and BCL-2 signalling in n-nitrosodiethylamine-induced murine hepatocellular carcinoma. Sci Rep. Dec 13 2019;9(1):19095. doi:10.1038/s41598-019-55666-x
  542. Coyne CP, Narayanan L. Mebendazole in simultaneous combination with dexamethasone-(C(21)-phosphoramidate)-[anti-EGFR] generated utilizing a novel synthesis regimen: dual anti-neoplastic cytotoxicity against pulmonary adenocarcinoma (A549). Journal of experimental therapeutics & oncology. Dec 2019;13(2):81-118.
  543. Williamson T, Mendes TB, Joe N, Cerutti JM, Riggins GJ. Mebendazole inhibits tumor growth and prevents lung metastasis in models of advanced thyroid cancer. Endocrine-related cancer. Mar 2020;27(3):123-136. doi:10.1530/erc-19-0341
  544. Zhang F, Li Y, Zhang H, et al. Anthelmintic mebendazole enhances cisplatin's effect on suppressing cell proliferation and promotes differentiation of head and neck squamous cell carcinoma (HNSCC). Oncotarget. Feb 21 2017;8(8):12968-12982. doi:10.18632/oncotarget.14673
  545. Sawanyawisuth K, Williamson T, Wongkham S, Riggins GJ. EFFECT OF THE ANTIPARASITIC DRUG MEBENDAZOLE ON CHOLANGIOCARCINOMA GROWTH. Southeast Asian J Trop Med Public Health. Nov 2014;45(6):1264-70.
  546. Martarelli D, Pompei P, Baldi C, Mazzoni G. Mebendazole inhibits growth of human adrenocortical carcinoma cell lines implanted in nude mice. Cancer chemotherapy and pharmacology. Apr 2008;61(5):809-17. doi:10.1007/s00280-007-0538-0
  547. Wang X, Lou K, Song X, et al. Mebendazole is a potent inhibitor to chemoresistant T cell acute lymphoblastic leukemia cells. Toxicology and applied pharmacology. Jun 1 2020;396:115001. doi:10.1016/j.taap.2020.115001
  548. Li Y, Thomas D, Deutzmann A, Majeti R, Felsher DW, Dill DL. Mebendazole for Differentiation Therapy of Acute Myeloid Leukemia Identified by a Lineage Maturation Index. Sci Rep. Nov 14 2019;9(1):16775. doi:10.1038/s41598-019-53290-3
  549. He L, Shi L, Du Z, et al. Mebendazole exhibits potent anti-leukemia activity on acute myeloid leukemia. Experimental cell research. Aug 1 2018;369(1):61-68. doi:10.1016/j.yexcr.2018.05.006
  550. Simbulan-Rosenthal CM, Dakshanamurthy S, Gaur A, et al. The repurposed anthelmintic mebendazole in combination with trametinib suppresses refractory NRASQ61K melanoma. Oncotarget. Feb 21 2017;8(8):12576-12595. doi:10.18632/oncotarget.14990
  551. Doudican N, Rodriguez A, Osman I, Orlow SJ. Mebendazole induces apoptosis via Bcl-2 inactivation in chemoresistant melanoma cells. Mol Cancer Res. Aug 2008;6(8):1308-15. doi:10.1158/1541-7786.Mcr-07-2159
  552. Chen XH, Xu YJ, Wang XG, et al. Mebendazole elicits potent antimyeloma activity by inhibiting the USP5/c-Maf axis. Acta Pharmacol Sin. Dec 2019;40(12):1568-1577. doi:10.1038/s41401-019-0249-1
  553. Skibinski CG, Williamson T, Riggins GJ. Mebendazole and radiation in combination increase survival through anticancer mechanisms in an intracranial rodent model of malignant meningioma. Journal of neuro-oncology. Dec 2018;140(3):529-538. doi:10.1007/s11060-018-03009-7
  554. Banovic P, Stankov S, Vranjes N, Zurkovic O, Capo I, Lalosevic D. Drug repurposing: mebendazole as effective antitumor agent. Are we seeing the whole story? Journal of BUON : official journal of the Balkan Union of Oncology . Nov-Dec 2018;23(6):1904-1911.
  555. Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-mebendazole as an anti-cancer agent. Ecancermedicalscience. 2014;8:443. doi:10.3332/ecancer.2014.443
  556. Dobrosotskaya IY, Hammer GD, Schteingart DE, Maturen KE, Worden FP. Mebendazole monotherapy and long-term disease control in metastatic adrenocortical carcinoma. Endocr Pract. May-Jun 2011;17(3):e59-62. doi:10.4158/ep10390.Cr
  557. Nygren P, Larsson R. Drug repositioning from bench to bedside: tumour remission by the antihelmintic drug mebendazole in refractory metastatic colon cancer. Acta oncologica (Stockholm, Sweden). Mar 2014;53(3):427-8. doi:10.3109/0284186x.2013.844359
  558. Patil VM, Bhelekar A, Menon N, et al. Reverse swing-M, phase 1 study of repurposing mebendazole in recurrent high-grade glioma. Cancer Med . Jul 2020;9(13):4676-4685. doi:10.1002/cam4.3094
  559. Giuli MV, Hanieh PN, Giuliani E, et al. Current Trends in ATRA Delivery for Cancer Therapy. Pharmaceutics. Jul 28 2020;12(8)doi:10.3390/pharmaceutics12080707
  560. Costantini L, Molinari R, Farinon B, Merendino N. Retinoic Acids in the Treatment of Most Lethal Solid Cancers. J Clin Med. Jan 28 2020;9(2)doi:10.3390/jcm9020360
  561. Ni X, Hu G, Cai X. The success and the challenge of all-trans retinoic acid in the treatment of cancer. Crit Rev Food Sci Nutr. 2019;59(sup1):S71-s80. doi:10.1080/10408398.2018.1509201
  562. Szymański Ł, Skopek R, Palusińska M, et al. Retinoic Acid and Its Derivatives in Skin. Cells. Dec 11 2020;9(12)doi:10.3390/cells9122660
  563. Uray IP, Dmitrovsky E, Brown PH. Retinoids and rexinoids in cancer prevention: from laboratory to clinic. Seminars in oncology. Feb 2016;43(1):49-64. doi:10.1053/j.seminoncol.2015.09.002
  564. de Oliveira MR. Vitamin A and Retinoids as Mitochondrial Toxicants. Oxid Med Cell Longev. 2015;2015:140267. doi:10.1155/2015/140267
  565. Xu A, Zhang N, Cao J, et al. Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation. Biochemical pharmacology. Jan 2020;171:113696. doi:10.1016/j.bcp.2019.113696
  566. Mezquita B, Mezquita C. Two Opposing Faces of Retinoic Acid: Induction of Stemness or Induction of Differentiation Depending on Cell-Type. Biomolecules. Oct 4 2019;9(10)doi:10.3390/biom9100567
  567. Schultze E, Collares T, Lucas CG, Seixas FK. Synergistic and additive effects of ATRA in combination with different anti-tumor compounds. Chem Biol Interact. Apr 1 2018;285:69-75. doi:10.1016/j.cbi.2018.02.021
  568. Lexicomp, Inc. All-trans retinoic acid (systemic tretinoin): Drug information. UpToDate. Accessed 01/19/2020, https://www.uptodate.com/contents/all-trans-retinoic-acid-systemic-tretinoin-drug-information. 2020;
  569. Mei D, Lv B, Chen B, et al. All-trans retinoic acid suppresses malignant characteristics of CD133-positive thyroid cancer stem cells and induces apoptosis. PLoS One. 2017;12(8):e0182835. doi:10.1371/journal.pone.0182835
  570. Cristiano MC, Cosco D, Celia C, et al. Anticancer activity of all-trans retinoic acid-loaded liposomes on human thyroid carcinoma cells. Colloids and surfaces B, Biointerfaces. Feb 1 2017;150:408-416. doi:10.1016/j.colsurfb.2016.10.052
  571. Malehmir M, Haghpanah V, Larijani B, et al. Multifaceted suppression of aggressive behavior of thyroid carcinoma by all-trans retinoic acid induced re-differentiation. Molecular and cellular endocrinology. Jan 2 2012;348(1):260-9. doi:10.1016/j.mce.2011.09.002
  572. Lan L, Cui D, Luo Y, et al. Inhibitory effects of retinoic acid on invasiveness of human thyroid carcinoma cell lines in vitro. Journal of endocrinological investigation. Oct 2009;32(9):731-8. doi:10.1007/bf03346528
  573. Garattini E, Bolis M, Garattini SK, et al. Retinoids and breast cancer: from basic studies to the clinic and back again. Cancer treatment reviews. Jul 2014;40(6):739-49. doi:10.1016/j.ctrv.2014.01.001
  574. Dou Y, Huang D, Zeng X, et al. All-trans retinoic acid enhances the effect of Fra-1 to inhibit cell proliferation and metabolism in cervical cancer. Biotechnology letters. Jun 2020;42(6):1051-1060. doi:10.1007/s10529-020-02847-8
  575. Guo JM, Xiao BX, Kang GZ, et al. Suppression of telomerase activity and arrest at G1 phase in human cervical cancer HeLa cells by all-trans retinoic acid. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society . Jan-Feb 2006;16(1):341-6. doi:10.1111/j.1525-1438.2006.00353.x
  576. Soprano KJ, Purev E, Vuocolo S, Soprano DR. Rb2/p130 and protein phosphatase 2A: key mediators of ovarian carcinoma cell growth suppression by all-trans retinoic acid. Oncogene. Aug 28 2006;25(38):5315-25. doi:10.1038/sj.onc.1209679
  577. Purev E, Soprano DR, Soprano KJ. Effect of all-trans retinoic acid on telomerase activity in ovarian cancer cells. Journal of experimental & clinical cancer research : CR. Jun 2004;23(2):309-16.
  578. Fang S, Hu C, Xu L, et al. All-trans-retinoic acid inhibits the malignant behaviors of hepatocarcinoma cells by regulating autophagy. Am J Transl Res. 2020;12(10):6793-6810.
  579. Zhao J, Wen G, Ding M, et al. Comparative proteomic analysis of colon cancer cell HCT-15 in response to all-trans retinoic acid treatment. Protein and peptide letters. Dec 2012;19(12):1272-80. doi:10.2174/092986612803521675
  580. Woo YJ, Jang KL. All-trans retinoic acid activates E-cadherin expression via promoter hypomethylation in the human colon carcinoma HCT116 cells. Biochemical and biophysical research communications. Sep 7 2012;425(4):944-9. doi:10.1016/j.bbrc.2012.08.038
  581. Zuo L, Yang X, Lu M, et al. All-Trans Retinoic Acid Inhibits Human Colorectal Cancer Cells RKO Migration via Downregulating Myosin Light Chain Kinase Expression through MAPK Signaling Pathway. Nutrition and cancer. Oct 2016;68(7):1225-33. doi:10.1080/01635581.2016.1216138
  582. Bouriez D, Giraud J, Gronnier C, Varon C. Efficiency of All-Trans Retinoic Acid on Gastric Cancer: A Narrative Literature Review. International journal of molecular sciences. Oct 29 2018;19(11)doi:10.3390/ijms19113388
  583. Kalemkerian GP, Jasti RK, Celano P, Nelkin BD, Mabry M. All-trans-retinoic acid alters myc gene expression and inhibits in vitro progression in small cell lung cancer. Cell Growth Differ. Jan 1994;5(1):55-60.
  584. Choi EJ, Whang YM, Kim SJ, Kim HJ, Kim YH. Combinational treatment with retinoic acid derivatives in non-small cell lung carcinoma in vitro. J Korean Med Sci. Sep 2007;22 Suppl(Suppl):S52-60. doi:10.3346/jkms.2007.22.S.S52
  585. Li J, Orr B, White K, et al. Chmp 1A is a mediator of the anti-proliferative effects of all-trans retinoic acid in human pancreatic cancer cells. Molecular cancer. Feb 12 2009;8:7. doi:10.1186/1476-4598-8-7
  586. Guo J, Xiao B, Lou Y, et al. Antitumor effects of all-trans-retinoic acid on cultured human pancreatic cancer cells. Journal of gastroenterology and hepatology. Feb 2006;21(2):443-8. doi:10.1111/j.1440-1746.2006.04180.x
  587. Albrechtsson E, Ohlsson B, Axelson J. The expression of retinoic acid receptors and the effects in vitro by retinoids in human pancreatic cancer cell lines. Pancreas. Jul 2002;25(1):49-56. doi:10.1097/00006676-200207000-00013
  588. Wang R, Liu C. All-trans retinoic acid therapy induces asymmetric division of glioma stem cells from the U87MG cell line. Oncology letters. Oct 2019;18(4):3646-3654. doi:10.3892/ol.2019.10691
  589. Li N, Lu Y, Li D, et al. All-trans retinoic acid suppresses the angiopoietin-Tie2 pathway and inhibits angiogenesis and metastasis in esophageal squamous cell carcinoma. PLoS One. 2017;12(4):e0174555. doi:10.1371/journal.pone.0174555
  590. Lu TY, Fan QX, Wang LX, Wang RL, Zhao PR, Lu SX. [Apoptosis in esophageal cancer cells induced by all-trans retinoic acid]. Zhonghua zhong liu za zhi [Chinese journal of oncology]. Nov 2007;29(11):822-5.
  591. Ortonne JP. Retinoic acid and pigment cells: a review of in-vitro and in-vivo studies. Br J Dermatol. Sep 1992;127 Suppl 41:43-7. doi:10.1111/j.1365-2133.1992.tb16987.x
  592. Zhang ML, Tao Y, Zhou WQ, et al. All-trans retinoic acid induces cell-cycle arrest in human cutaneous squamous carcinoma cells by inhibiting the mitogen-activated protein kinase-activated protein 1 pathway. Clin Exp Dermatol. Apr 2014;39(3):354-60. doi:10.1111/ced.12227
  593. Kunisada M, Budiyanto A, Bito T, Nishigori C, Ueda M. Retinoic acid suppresses telomerase activity in HSC-1 human cutaneous squamous cell carcinoma. Br J Dermatol. Mar 2005;152(3):435-43. doi:10.1111/j.1365-2133.2005.06471x
  594. Rudkin GH, Carlsen BT, Chung CY, et al. Retinoids inhibit squamous cell carcinoma growth and intercellular communication. The Journal of surgical research. Apr 2002;103(2):183-9. doi:10.1006/jsre.2001.6346
  595. Takahashi S. Current Understandings of Myeloid Differentiation Inducers in Leukemia Therapy. Acta haematologica. Nov 20 2020:1-9. doi:10.1159/000510980
  596. Barna G, Sebestyén A, Weischede S, et al. Different ways to induce apoptosis by fenretinide and all-trans-retinoic acid in human B lymphoma cells. Anticancer research. Nov-Dec 2005;25(6b):4179-85.
  597. Niitsu N, Higashihara M, Honma Y. Human B-cell lymphoma cell lines are highly sensitive to apoptosis induced by all-trans retinoic acid and interferon-gamma. Leuk Res. Aug 2002;26(8):745-55. doi:10.1016/s0145-2126(01)00202-8
  598. Bonnefoix T, Gressin R, Jacrot M, et al. Growth modulation of freshly isolated non-Hodgkin's B-lymphoma cells induced by various cytokines and all-trans-retinoic-acid. Leuk Lymphoma. Mar 1997;25(1-2):169-78. doi:10.3109/10428199709042507
  599. Traylor JI, Sheppard HE, Ravikumar V, et al. Computational Drug Repositioning Identifies Potentially Active Therapies for Chordoma. Neurosurgery. Jan 13 2021;88(2):428-436. doi:10.1093/neuros/nyaa398
  600. Bayrak OF, Aydemir E, Gulluoglu S, et al. The effects of chemotherapeutic agents on differentiated chordoma cells. J Neurosurg Spine. Dec 2011;15(6):620-4. doi:10.3171/2011.7.Spine10798
  601. NIH. National Institutes of Health. Clinical Trials.Gov. Available at https://clinicaltrials.gov/ct2/results?term=ATRA&cond=cancer&recrs=b&recrs=a&recrs=f&recrs=d&age_v=&gndr=&type=Intr&rslt=&Search=Apply . Accessed 02/04/21. 2021;
  602. StatPearls. Medical Reference: Acute Promyelocytic Leukemia. Updated 2021. Accessed 2/9/2021, https://www.statpearls.com/articlelibrary/viewarticle/27807/
  603. Rossetti S, Sacchi N. Emerging Cancer Epigenetic Mechanisms Regulated by All-Trans Retinoic Acid. Cancers. Aug 14 2020;12(8)doi:10.3390/cancers12082275
  604. Pan P, Chen X. Nuclear Receptors as Potential Therapeutic Targets for Myeloid Leukemia. Cells. Aug 19 2020;9(9)doi:10.3390/cells9091921
  605. Dobrotkova V, Chlapek P, Jezova M, et al. Prediction of neuroblastoma cell response to treatment with natural or synthetic retinoids using selected protein biomarkers. PLOS ONE. 2019;14(6):e0218269. doi:10.1371/journal.pone.0218269
  606. Shohet JM, Lowas SR, Nuchtern JG. Treatment and prognosis of neuroblastoma. UpToDate. Updated 12/15/2020. Accessed 2/9/2021, https://www.uptodate.com/contents/treatment-and-prognosis-of-neuroblastoma
  607. Schlenk RF, Fröhling S, Hartmann F, et al. Phase III study of all-trans retinoic acid in previously untreated patients 61 years or older with acute myeloid leukemia. Leukemia. Nov 2004;18(11):1798-803. doi:10.1038/sj.leu.2403528
  608. Ferrero D, Crisà E, Marmont F, et al. Survival improvement of poor-prognosis AML/MDS patients by maintenance treatment with low-dose chemotherapy and differentiating agents. Annals of hematology. Aug 2014;93(8):1391-400. doi:10.1007/s00277-014-2047-7
  609. Soriano AO, Yang H, Faderl S, et al. Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood. Oct 1 2007;110(7):2302-8. doi:10.1182/blood-2007-03-078576
  610. Kuendgen A, Knipp S, Fox F, et al. Results of a phase 2 study of valproic acid alone or in combination with all-trans retinoic acid in 75 patients with myelodysplastic syndrome and relapsed or refractory acute myeloid leukemia. Annals of hematology. Dec 2005;84 Suppl 1:61-6. doi:10.1007/s00277-005-0026-8
  611. Estey EH, Thall PF, Pierce S, et al. Randomized phase II study of fludarabine + cytosine arabinoside + idarubicin +/- all-trans retinoic acid +/- granulocyte colony-stimulating factor in poor prognosis newly diagnosed acute myeloid leukemia and myelodysplastic syndrome. Blood. Apr 15 1999;93(8):2478-84.
  612. Bug G, Ritter M, Wassmann B, et al. Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer. Dec 15 2005;104(12):2717-25. doi:10.1002/cncr.21589
  613. Pilatrino C, Cilloni D, Messa E, et al. Increase in platelet count in older, poor-risk patients with acute myeloid leukemia or myelodysplastic syndrome treated with valproic acid and all-trans retinoic acid. Cancer. Jul 1 2005;104(1):101-9. doi:10.1002/cncr.21132
  614. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. Mar 15 2007;109(6):1114-24. doi:10.1002/cncr.22496
  615. Küley-Bagheri Y, Kreuzer KA, Monsef I, Lübbert M, Skoetz N. Effects of all-trans retinoic acid (ATRA) in addition to chemotherapy for adults with acute myeloid leukaemia (AML) (non-acute promyelocytic leukaemia (non-APL)). The Cochrane database of systematic reviews. Aug 6 2018;8(8):Cd011960. doi:10.1002/14651858.CD011960.pub2
  616. Smith MA, Adamson PC, Balis FM, et al. Phase I and pharmacokinetic evaluation of all-trans-retinoic acid in pediatric patients with cancer. J Clin Oncol. Nov 1992;10(11):1666-73. doi:10.1200/jco.1992.10.11.1666
  617. Kocher HM, Basu B, Froeling FEM, et al. Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer. Nature communications. Sep 24 2020;11(1):4841. doi:10.1038/s41467-020-18636-w
  618. Zhang Y, Jia S, Liu Y, et al. A clinical study of all-trans-retinoid-induced differentiation therapy of advanced thyroid cancer. Nuclear medicine communications. Apr 2007;28(4):251-5. doi:10.1097/MNM.0b013e3280708ebf
  619. Meyskens FL, Jr., Surwit E, Moon TE, et al. Enhancement of regression of cervical intraepithelial neoplasia II (moderate dysplasia) with topically applied all-trans-retinoic acid: a randomized trial. Journal of the National Cancer Institute. Apr 6 1994;86(7):539-43. doi:10.1093/jnci/86.7.539
  620. Ruffin MT, Bailey JM, Normolle DP, et al. Low-dose topical delivery of all-trans retinoic acid for cervical intraepithelial neoplasia II and III. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology . Dec 2004;13(12):2148-52.
  621. Braud AC, Gonzague L, Bertucci F, et al. Retinoids, cisplatin and interferon-alpha in recurrent or metastatic cervical squamous cell carcinoma: clinical results of 2 phase II trials. European cytokine network. Jan-Mar 2002;13(1):115-20.
  622. Goncalves A, Camerlo J, Bun H, et al. Phase II study of a combination of cisplatin, all-trans-retinoic acid and interferon-alpha in squamous cell carcinoma: clinical results and pharmacokinetics. Anticancer research. Mar-Apr 2001;21(2b):1431-7.
  623. Bryan M, Pulte ED, Toomey KC, et al. A pilot phase II trial of all-trans retinoic acid (Vesanoid) and paclitaxel (Taxol) in patients with recurrent or metastatic breast cancer. Investigational new drugs. Dec 2011;29(6):1482-7. doi:10.1007/s10637-010-9478-3
  624. Sutton LM, Warmuth MA, Petros WP, Winer EP. Pharmacokinetics and clinical impact of all-trans retinoic acid in metastatic breast cancer: a phase II trial. Cancer chemotherapy and pharmacology. 1997;40(4):335-41. doi:10.1007/s002800050666
  625. Budd GT, Adamson PC, Gupta M, et al. Phase I/II trial of all-trans retinoic acid and tamoxifen in patients with advanced breast cancer. Clin Cancer Res. Mar 1998;4(3):635-42.
  626. Falanga A, Toma S, Marchetti M, et al. Effect of all-trans-retinoic acid on the hypercoagulable state of patients with breast cancer. Am J Hematol. May 2002;70(1):9-15. doi:10.1002/ajh.10073
  627. Treat J, Greenberg R, Bratschi J, Gorman G, Meehan L, Friedland D. First hints in non-small cell lung cancer (NSCLC). Leukemia. 1994;8 Suppl 3:S55-8.
  628. Treat J, Friedland D, Luginbuhl W, et al. Phase II trial of all-trans retinoic acid in metastatic non-small cell lung cancer. Cancer investigation. 1996;14(5):415-20. doi:10.3109/07357909609018898
  629. Thiruvengadam R, Atiba JO, Azawi SH. A phase II trial of a differentiating agent (tRA) with cisplatin-VP 16 chemotherapy in advanced non-small cell lung cancer. Investigational new drugs. 1996;14(4):395-401. doi:10.1007/bf00180817
  630. Athanasiadis I, Kies MS, Miller M, et al. Phase II study of all-trans-retinoic acid and alpha-interferon in patients with advanced non-small cell lung cancer. Clin Cancer Res. Sep 1995;1(9):973-9.
  631. Arrieta O, González-De la Rosa CH, Aréchaga-Ocampo E, et al. Randomized phase II trial of All-trans-retinoic acid with chemotherapy based on paclitaxel and cisplatin as first-line treatment in patients with advanced non-small-cell lung cancer. J Clin Oncol. Jul 20 2010;28(21):3463-71. doi:10.1200/jco.2009.26.6452
  632. Kalemkerian GP, Jiroutek M, Ettinger DS, Dorighi JA, Johnson DH, Mabry M. A phase II study of all-trans-retinoic acid plus cisplatin and etoposide in patients with extensive stage small cell lung carcinoma: an Eastern Cooperative Oncology Group Study. Cancer. Sep 15 1998;83(6):1102-8. doi:10.1002/(sici)1097-0142(19980915)83:6<1102::aid-cncr8>3.0.co;2-9
  633. Arrieta Ó, Hernández-Pedro N, Fernández-González-Aragón MC, et al. Retinoic acid reduces chemotherapy-induced neuropathy in an animal model and patients with lung cancer. Neurology. Sep 6 2011;77(10):987-95. doi:10.1212/WNL.0b013e31822e045c
  634. Tobin RP, Jordan KR, Robinson WA, et al. Targeting myeloid-derived suppressor cells using all-trans retinoic acid in melanoma patients treated with Ipilimumab. International immunopharmacology. Oct 2018;63:282-291. doi:10.1016/j.intimp.2018.08.007
  635. Sondak VK, Liu PY, Flaherty LE, et al. A phase II evaluation of all-trans-retinoic acid plus interferon alfa-2a in stage IV melanoma: a Southwest Oncology Group study. Cancer J Sci Am. Jan-Feb 1999;5(1):41-7.
  636. Meyskens FL, Jr., Edwards L, Levine NS. Role of topical tretinoin in melanoma and dysplastic nevi. J Am Acad Dermatol. Oct 1986;15(4 Pt 2):822-5. doi:10.1016/s0190-9622(86)70239-9
  637. Halpern AC, Schuchter LM, Elder DE, et al. Effects of topical tretinoin on dysplastic nevi. J Clin Oncol. May 1994;12(5):1028-35. doi:10.1200/jco.1994.12.5.1028
  638. Stam-Posthuma JJ, Vink J, le Cessie S, Bruijn JA, Bergman W, Pavel S. Effect of topical tretinoin under occlusion on atypical naevi. Melanoma research. Dec 1998;8(6):539-48. doi:10.1097/00008390-199812000-00009
  639. Bettoli V, Zauli S, Virgili A. Retinoids in the chemoprevention of non-melanoma skin cancers: why, when and how. The Journal of dermatological treatment. Jun 2013;24(3):235-7. doi:10.3109/09546634.2012.746634
  640. Fayne R, Nanda S, Nichols A, Shen J. Combination Topical Chemotherapy for the Treatment of an Invasive Cutaneous Squamous Cell Carcinoma. J Drugs Dermatol. Feb 1 2020;19(2):202-204. doi:10.36849/jdd.2020.2228
  641. Ianhez M, Fleury LF, Jr., Miot HA, Bagatin E. Retinoids for prevention and treatment of actinic keratosis. Anais brasileiros de dermatologia. Jul-Aug 2013;88(4):585-93. doi:10.1590/abd1806-4841.20131803
  642. Weinstock MA, Bingham SF, Digiovanna JJ, et al. Tretinoin and the prevention of keratinocyte carcinoma (Basal and squamous cell carcinoma of the skin): a veterans affairs randomized chemoprevention trial. J Invest Dermatol. Jun 2012;132(6):1583-90. doi:10.1038/jid.2011.483
  643. Weinstock MA, Bingham SF, Lew RA, et al. Topical tretinoin therapy and all-cause mortality. Arch Dermatol. Jan 2009;145(1):18-24. doi:10.1001/archdermatol.2008.542
  644. Clerici C, Castellani D, Russo G, et al. Treatment with all-trans retinoic acid plus tamoxifen and vitamin E in advanced hepatocellular carcinoma. Anticancer research. Mar-Apr 2004;24(2c):1255-60.
  645. Meyskens FL, Jr., Jacobson J, Nguyen B, Weiss GR, Gandara DR, MacDonald JS. Phase II trial of oral beta-all trans-retinoic acid in hepatocellular carcinoma (SWOG 9157). Investigational new drugs. 1998;16(2):171-3. doi:10.1023/a:1006032706362
  646. Jin J, Li X, Xing L, et al. Addition of all-trans-retinoic acid to omeprazole and sucralfate therapy improves the prognosis of gastric dysplasia. The Journal of international medical research. Apr 2015;43(2):204-16. doi:10.1177/0300060514559791
  647. Goldberg JS, Vargas M, Rosmarin AS, et al. Phase I trial of interferon alpha2b and liposome-encapsulated all-trans retinoic acid in the treatment of patients with advanced renal cell carcinoma. Cancer. Sep 15 2002;95(6):1220-7. doi:10.1002/cncr.10809
  648. Schenk T, Stengel S, Zelent A. Unlocking the potential of retinoic acid in anticancer therapy. British journal of cancer. 2014;111(11):2039-2045. doi:10.1038/bjc.2014.412
  649. Geng A, Weinstock MA, Hall R, Eilers D, Naylor M, Kalivas J. Tolerability of high-dose topical tretinoin: the Veterans Affairs Topical Tretinoin Chemoprevention Trial. Br J Dermatol. Oct 2009;161(4):918-24. doi:10.1111/j.1365-2133.2009.09341.x
  650. Lytras T, Nikolopoulos G, Bonovas S. Statins and the risk of colorectal cancer: an updated systematic review and meta-analysis of 40 studies. World J Gastroenterol. Feb 21 2014;20(7):1858-70. doi:10.3748/wjg.v20.i7.1858
  651. Han KT, Kim S. Do cholesterol levels and continuity of statin use affect colorectal cancer incidence in older adults under 75 years of age? PLoS One. 2021;16(4):e0250716. doi:10.1371/journal.pone.0250716
  652. Ling Y, Yang L, Huang H, et al. Prognostic Significance of Statin Use in Colorectal Cancer: A Systematic Review and Meta-Analysis. Medicine. Jun 2015;94(25):e908. doi:10.1097/md.0000000000000908
  653. Lash TL, Riis AH, Ostenfeld EB, et al. Associations of Statin Use With Colorectal Cancer Recurrence and Mortality in a Danish Cohort. American journal of epidemiology. Sep 15 2017;186(6):679-687. doi:10.1093/aje/kww245
  654. Macrae FA. Colorectal cancer: Epidemiology, risk factors, and protective factors. UpToDate. Updated 2/17/2021. Accessed 6/1/2021, https://www.uptodate.com/contents/colorectal-cancer-epidemiology-risk-factors-and-protective-factors
  655. Qi JH, Wei JN, Zhang ZJ, et al. [A Meta-analysis on association between statins and colorectal cancer]. Zhonghua liu xing bing xue za zhi = Zhonghua liuxingbingxue zazhi. Feb 10 2021;42(2):343-350. doi:10.3760/cma.j.cn112338-20200119-00045
  656. Pourlotfi A, Ahl R, Sjolin G, et al. Statin therapy and postoperative short-term mortality after rectal cancer surgery. https://doi.org/10.1111/codi.15481. Colorectal Disease. 2021/04/01 2021;23(4):875-881. doi:https://doi.org/10.1111/codi.15481
  657. Liang PS, Shaukat A, Crockett SD. AGA Clinical Practice Update on Chemoprevention for Colorectal Neoplasia: Expert Review. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. Feb 10 2021;doi:10.1016/j.cgh.2021.02.014
  658. Sonnenblick A, Agbor-Tarh D, Bradbury I, et al. Impact of Diabetes, Insulin, and Metformin Use on the Outcome of Patients With Human Epidermal Growth Factor Receptor 2–Positive Primary Breast Cancer: Analysis From the ALTTO Phase III Randomized Trial. Journal of Clinical Oncology. 2017;35(13):1421-1429. doi:10.1200/jco.2016.69.7722.
  659. Skuli SJ, Alomari S, Gaitsch H, Bakayoko A, Skuli N, Tyler BM. Metformin and Cancer, an Ambiguanidous Relationship. Pharmaceuticals (Basel). May 19 2022;15(5)doi:10.3390/ph15050626. https://mdpi-res.com/d_attachment/pharmaceuticals/pharmaceuticals-15-00626/article_deploy/pharmaceuticals-15-00626-v2.pdf?version=1653014705
  660. Cejuela M, Martin-Castillo B, Menendez JA, Pernas S. Metformin and Breast Cancer: Where Are We Now? Int J Mol Sci. Feb 28 2022;23(5)doi:10.3390/ijms23052705. https://mdpi-res.com/d_attachment/ijms/ijms-23-02705/article_deploy/ijms-23-02705-v3.pdf?version=1646269562
  661. Goodwin PJ, Chen BE, Gelmon KA, et al. Effect of Metformin vs Placebo on Invasive Disease-Free Survival in Patients With Breast Cancer: The MA.32 Randomized Clinical Trial. Jama. May 24 2022;327(20):1963-1973. doi:10.1001/jama.2022.6147. https://jamanetwork.com/journals/jama/articlepdf/2792615/jama_goodwin_2022_oi_220046_1652813403.66253.pdf
  662. Morio K, Kurata Y, Kawaguchi-Sakita N, Shiroshita A, Kataoka Y. Efficacy of Metformin in Patients With Breast Cancer Receiving Chemotherapy or Endocrine Therapy: Systematic Review and Meta-analysis. Ann Pharmacother. Mar 2022;56(3):245-255. doi:10.1177/10600280211025792. https://journals.sagepub.com/doi/10.1177/10600280211025792?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dpubmed
  663. Wang Q, Ma X, Long J, Du X, Pan B, Mao H. Metformin and survival of women with breast cancer: A meta-analysis of randomized controlled trials. J Clin Pharm Ther. Mar 2022;47(3):263-269. doi:10.1111/jcpt.13500. https://onlinelibrary.wiley.com/doi/pdfdirect/10.1111/jcpt.13500?download=true
  664. Markowska A, Kaysiewicz J, Markowska J, Huczyński A. Doxycycline, salinomycin, monensin and ivermectin repositioned as cancer drugs. Bioorganic & medicinal chemistry letters. Jul 1 2019;29(13):1549-1554. doi:10.1016/j.bmcl.2019.04.045.
  665. Sun T, Zhao N, Ni C-s, et al. Doxycycline inhibits the adhesion and migration of melanoma cells by inhibiting the expression and phosphorylation of focal adhesion kinase (FAK). Cancer Letters. 2009/11/28/ 2009;285(2):141-150. doi:https://doi.org/10.1016/j.canlet.2009.05.004. https://www.sciencedirect.com/science/article/pii/S0304383509003425
  666. Zhong W, Chen S, Qin Y, et al. Doxycycline inhibits breast cancer EMT and metastasis through PAR-1/NF-κB/miR-17/E-cadherin pathway. Oncotarget. Dec 1 2017;8(62):104855-104866. doi:10.18632/oncotarget.20418.
  667. Patel RS, Parmar M. Doxycycline Hyclate. StatPearls. StatPearls Publishing Copyright © 2022, StatPearls Publishing LLC.; 2022.
  668. Schwartz E. Appendix A: Drugs for Parasitic Infections. Tropical Diseases in Travelers. 2009:436-465.
  669. Shen L-C, Chen Y-K, Lin L-M, Shaw S-Y. Anti-invasion and anti-tumor growth effect of doxycycline treatment for human oral squamous-cell carcinoma – In vitro and in vivo studies. Oral Oncology. 2010/03/01/ 2010;46(3):178-184. doi:https://doi.org/10.1016/j.oraloncology.2009.11.013. https://www.sciencedirect.com/science/article/pii/S1368837509009828
  670. Tang X, Wang X, Zhao YY, Curtis JM, Brindley DN. Doxycycline attenuates breast cancer related inflammation by decreasing plasma lysophosphatidate concentrations and inhibiting NF-κB activation. Mol Cancer. Feb 8 2017;16(1):36. doi:10.1186/s12943-017-0607-x.
  671. Delaunay S, Pascual G, Feng B, et al. Mitochondrial RNA modifications shape metabolic plasticity in metastasis. Nature. 2022/06/29 2022;doi:10.1038/s41586-022-04898-5.
    https://www.nature.com/articles/s41586-022-04898-5.pdf
  672. Ottawa Hospital Research Institute. Combining Doxycycline With Bone-Targeted Therapy in Patients With Metastatic Breast Cancer (OTT12-05). Accessed July 6, 2022, https://clinicaltrials.gov/ct2/show/NCT01847976
  673. Sidney Kimmel Cancer Center at Thomas Jefferson University. Metformin Hydrochloride and Doxycycline in Treating Patients With Localized Breast or Uterine Cancer. Accessed July 6, 2022, https://clinicaltrials.gov/ct2/show/NCT02874430
  674. Medical College of Wisconsin. Efficacy of Doxycycline on Metakaryote Cell Death in Patients With Resectable Pancreatic Cancer. Accessed July 6, 2022, https://clinicaltrials.gov/ct2/show/NCT02775695
  675. Walcher L, Kistenmacher AK, Suo H, et al. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front Immunol. 2020;11:1280. doi:10.3389/fimmu.2020.01280. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7426526/pdf/fimmu-11-01280.pdf
  676. Scatena C, Roncella M, Di Paolo A, et al. Doxycycline, an Inhibitor of Mitochondrial Biogenesis, Effectively Reduces Cancer Stem Cells (CSCs) in Early Breast Cancer Patients: A Clinical Pilot Study. Frontiers in oncology. 2018;8:452. doi:10.3389/fonc.2018.00452. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6194352/pdf/fonc-08-00452.pdf
  677. Chanprapaph K, Vachiramon V, Rattanakaemakorn P. Epidermal growth factor receptor inhibitors: a review of cutaneous adverse events and management. Dermatol Res Pract. 2014;2014:734249. doi:10.1155/2014/734249. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3958662/pdf/DRP2014-734249.pdf
  678. Raimondi A, Corallo S, Lonardi S, et al. Systemic doxycycline for pre-emptive treatment of anti-EGFR-related skin toxicity in patients with metastatic colorectal cancer receiving first-line panitumumab-based therapy: a post hoc analysis of the Valentino study. Support Care Cancer. Jul 2021;29(7):3971-3980. doi:10.1007/s00520-020-05972-2.
  679. Ramírez-Daffós P, Jiménez-Orozco E, Bolaños M, et al. A phase 2 study for evaluating doxycycline 50 mg once daily and 100 mg once daily as preemptive treatment for skin toxicity in patients with metastatic colorectal cancer treated with an anti-EGFR and chemotherapy. Support Care Cancer. Jul 1 2022;doi:10.1007/s00520-022-07254-5.
  680. Deplanque G, Gervais R, Vergnenegre A, et al. Doxycycline for prevention of erlotinib-induced rash in patients with non-small-cell lung cancer (NSCLC) after failure of first-line chemotherapy: A randomized, open-label trial. J Am Acad Dermatol. Jun 2016;74(6):1077-85. doi:10.1016/j.jaad.2016.01.019.
  681. Kuzdzał J, Sładek K, Wasowski D, et al. Talc powder vs doxycycline in the control of malignant pleural effusion: a prospective, randomized trial. Med Sci Monit. Jun 2003;9(6):Pi54-9.
  682. Rafiei R, Yazdani B, Ranjbar SM, et al. Long-term results of pleurodesis in malignant pleural effusions: Doxycycline vs Bleomycin. Advanced biomedical research. 2014;3:149. doi:10.4103/2277-9175.137831.
  683. Bonnetblanc JM. [Doxycycline]. Annales de dermatologie et de venereologie. Jun-Jul 2002;129(6-7):874-82. Doxycycline.
  684. Jung WJ, Jang S, Choi WJ, et al. Metformin administration is associated with enhanced response to transarterial chemoembolization for hepatocellular carcinoma in type 2 diabetes patients. Scientific Reports. 2022/08/25 2022;12(1):14482. doi:10.1038/s41598-022-18341-2