Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2021 Mar 1;11(3):884–897.

The role of the renin-angiotensin system inhibitors in malignancy: a review

Ju Yang 1,*, Xi Yang 2,*, Ling Gao 2,*, Jie Zhang 3, Cheng Yi 2, Ying Huang 1
PMCID: PMC7994166  PMID: 33791161

Abstract

Hypertension is one of the most prevalent diseases in cardiology. The angiotensin receptor blockers (ARBs)/angiotensin converting enzyme inhibitors (ACEIs) are widely used drugs to stabilize the blood pressure via inhibition of the renin-angiotensin system (RAS). Studies have found that the exposure to RAS inhibitors (RASi) can suppress the development of cancers via multimodal mechanisms and has attracted increased attentions in the recent past. Owing the potential of RASi to inhibit tumor growth, proliferation and metastasis, they are considered as the potential and exciting candidates to enhance the effect of chemo-radiotherapy and targeted therapy efficacy. However, there are conflicting reports as to the use of ARB/ACEI in all facets of tumor growth. In this study, we comprehensively summarize and review the potential mechanisms of RASi in cancer treatment, like inhibition of tumor angiogenesis, reduction of cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM), regulation of immune cells and improvement of hypoxia. Additionally, based on the basic and clinical experiments, we analyze the views and results regarding the role of RASi plays in tumor from genesis to recurrence, and certainly cancer treatment (chemo-radiotherapy and targeted therapy). In the last, not only do we discuss the prospects of using RASi to enhance cancer treatment efficacy but also point out the conflicting situation with the intention to give some directions and inspiration on this topic.

Keywords: RASi, ARB, AECI, cancer treatment

Introduction

Cancer has been one of the devastating human diseases that threatens human health and researchers across the globe are directing studies for the development of cancer treatment. According to cancer statistics, in the United States 1,806,590 new cancer cases and 606,520 cancer related deaths are estimated to occur in 2020 [1]. Chemo-radiotherapy and targeted therapy are part of the established standard treatment strategies employed for the management of human cancers. Chemotherapy is one major treatment for malignancy, but the actual effects and responses to it are confined to drug distributions and immune milieu in tumor [2]. Similarly, radiotherapy efficacy has also been confronted with the resistance caused by tumor hypoxia [3]. While as one common targeted drug-tyrosine kinase inhibitors have also raised people’s concerns due to its cardiovascular toxicity, like hypertension, heart failure and arrhythmia [4,5]. Besides, researchers have discovered that the abnormal fibroblasts, extracellular matrix (ECM), and immune cells associated with tumors are all adverse to tumor microenvironment (TME), which on one hand accelerates the tumor progression and also impacts the effects of all kinds of anticancer therapies [6]. In this case, it is of highly importance to develop a strategy to improve anticancer effects and mitigate cardiovascular toxicity [2,7,8].

The renin-angiotensin system (RAS) is well-known for its role in maintaining cardiovascular homeostasis and electrolyte balance [9]. There are two main RAS inhibitors (RASi), angiotensin receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs). Both of these are widely used in the treatment of cardiovascular and renal disease [10-13]. ACEIs are known to inhibit synthesis of angiotensin II (Ang II), which exerts its cellular role predominantly by the angiotensin II receptor type 1 (AT1R). While ARBs selectively hinder its binding to downstream angiotensin receptors as shown in Figure 1 [14]. Increased vascular wall thickness and vasoconstriction which are related to systematic or endocrine signaling of the RAS are both the critical factors for the distal metastasis of tumor cells [15]. Recently, attention has been shifted to its relationship with cancer. Many reports have suggested there exists an association between RASi and different aspects of tumor genesis, for instance, they may decrease the expression of the vascular endothelial growth factor (VEGF), enhance drug delivery, reduce the interstitial extracellular matrix (ECM) content so improve hypoxia and regulate TME [9,16-18]. Thus, these multimodal mechanisms provide a basis for the potential applicability of the application of RASi in cancer treatment.

Figure 1.

Figure 1

The conventional and identified pathways of RAS axis. Angiotensinogen (AGT) is produced by the liver, then hydrolyzed by renin. Renin, produced by the juxtaglomerular cells of the kidney and then form Angiotensin I (Ang I). Next, Ang I is hydrolyzed by angiotensin-converting enzyme (ACE), to produce the downstream receptor-angiotensin II (Ang II). Ang II usually responses to two receptors, angiotensin II type 1 receptor (AT1R) and angiotensin II type 2 receptor (AT2R). ACE2 catalyzes Ang II to Angiotensin (1-7) (Ang (1-7)), which are also the product of cleaving Angiotensin (1-9) (Ang (1-9)). Ang (1-7) commonly interact with mitochondrial assembly receptor (Mas receptor or MasR). Cleavaged by ACE and decarboxylase (DC) respectively, Ang A and Ang (1-7) was connected through Alamandine, which siganals through MbbAS-related G protein couple receptor D (MRGD). In addition to Ang II, there are Angiotensin III (Ang III) and Angiotensin IV (Ang IV) identified in this path. IRAP (insulin-regulated membrane aminopeptidase; also called AT4R) is a binding site for Ang IV (1-7). APA, aminopeptidase A; APN, aminopep-tidase N; DC, decarboxylase; MLDAD, mononuclear leukocyte-derived aspartate DC; NEP, neutral endopeptidase; PEP, prolyendopeptidase [9].

Indeed, more researchers and medical institutions have examined and confirmed the efficacy of implementing these strategies. Take losartan (one common drug of the ARBs) for example, researchers have found its role in reducing tumor growth rates in human pancreatic, breast and prostate cancer cell xenografts [2,19,20]. Similarly, captopril (one ACEI) not only inhibited tumor angiogenesis but also reduced the extent of liver metastases [21]. Given this background, the purpose of the present review was, therefore, to discuss the controversial effects of RAS blockade in experimental models and clinical practice. Nonetheless, whether RAS blockade has positive role in the incidence and intervention of cancer remains a topic of debate [22,23]. Herein, we summarize such studies and their results in this essay. Although there have already been reviews that discussed this topic, but here we comprehensively and lately reviewed the potential mechanisms, which include but are not limited to drug distribution and TME, from the whole process of cancer (genesis, treatment and recurrence) analyzed supportive and negative results or views so far about the role of RASi in cancers. Finally, we discuss the possible directions to current controversial points related to this subject.

The mechanisms of RASi’s potential therapeutic possibilities in tumor

Tumor angiogenesis and AT1R

Angiogenesis is one of the key hallmarks of cancer and is regulated by numerous growth factors, among which VEGF may be the most crucial one in tumor angiogenesis [24-26]. Consistently, a number of studies have found that ARB can inhibit the proliferation of pancreatic cancer cell lines by inhibiting VEGF-mediated angiogenesis in tumors [9,27]. Besides, reduced vessel leakiness by inhibiting VEGF has shown to induce structurally leaky and hyper-permeability, which could also prevent tumor hypoxia and acidosis. One certain thing is that RASi can decompress tumor blood vessels to enhance drug and oxygen delivery. For example, results from the drug losartan showed a significant improvement in the perfused vessel fraction, both in breast and pancreatic tumors [2]. In addition, Zhao et al also found that losartan improved perfusion and attenuated tumor hypoxia. In this report, they measured the fraction of perfused vessels and the level of tumor hypoxia by immune histology [28]. The results showed that the percentage of perfused blood vessels in losartan group greatly surpassed the control group. It has suggested that Ang II/AT1R may promote VEGF secretion and expression in various cancers [29,30]. Moreover, about the two prominent angiotensin receptors-AT1R and angiotensin II type 2 receptor (AT2R), so far there also have been conflicting reports on them. Some studies implied a negative growth regulatory function for AT1R, wherein it has been found that Agtrα1 gene (encoded AT1R) is down-regulated in melanoma cell lines by methylating CpG island. Conversely Agtr2 gene (encoded AT2R) was over-expressed in 75% melanoma brain metastatic cell lines. Other studies have suggested the expression of AGT1R is much higher than AGT2R in breast and pancreatic tumor cells. This implies that the role of AT1R and AT2R is possibly cell type specific and context dependent [31-33].

Cancer-associated fibroblasts (CAFs)

Fibroblasts are spindle-shaped cells that synthesize collagen in connective tissues [34]. In normal tissues, located in the ECM, fibroblasts are activated during the process of wound healing, tissue inflammation and organ fibrosis [35]. Such activated fibroblasts associated with cancer are called as CAFs or myofibroblasts [36]. Generally, CAFs are indispensable in shaping the TME to promote tumor growth through the production of multiple ECM proteins and diverse interactions with cancer and immune cells. Consequently, CAFs interfere with tumor immunity, metabolic reprogramming of the TME, ECM remodeling, and efficacy of therapies [34,37]. Several studies have demonstrated that by blocking Ang II signaling and targeting AT1R, losartan reprogrammed the state of CAFs-from active to quiescent, and reduced the level of α-smooth-muscle actin (α-SMA, as the marker of activated CAF) [2,38].

Solid stress and ECM content

Solid stress is a result of the physical forces generated by ECM and cancer cells in the TME [39,40]. Therefore, lowering the levels of crucial components of the ECM-collagen and hyaluronic acid is a valid approach to anticancer treatments, which has been strongly confirmed by several reports. Zhao et al reported that the number of intra-tumoral α-SMA positive stromal cells decreased significantly thus also reduced ECM content after the use of losartan. When the solid stress levels were assessed, the losartan treatment group showed lower levels than the control group, regardless of compressive stress or tensile stress [28]. The same effect was suggested in a report that tested a therapy targeting both collagen and hyaluronan. Moreover, it turned out that losartan reduced collagen I production in CAFs. In two cell lines tested (pancreatic and breast tumors), the solid stress of control group was double that of the losartan group, which suggests that the treatment, via its anti-fibrotic effects, could possibly reduce the tumor solid stress [2].

Macrophage

Available studies have demonstrated that Ang II/AT1R axis is of great importance in the maturation and function of immune-stimulatory myeloid cells [9]. Similarly, multiple studies have reported that the Ang II can stimulate the release and infiltration of macrophage/monocyte chemo-attractant protein. This has been shown to accelerate cancer progression and correlates significantly with high prostate-specific antigen (PSA) that is linked to recurrence rates in human prostate cancer [41-44]. Meanwhile, studies have pointed that the use of ARB could down-regulate monocyte chemoattractant protein (MCP)-1 expression and macrophage infiltration in prostate cancer. In contrast, the effects of ACE on macrophages seem to be independent of Ang II/AT1R axis [45]. One study suggested that in vitro overexpressed ACE resulted in the myeloid maturation reduction of myeloid-derived suppressor cells (MDSCs), which are believed to inhibit T-cell activity and immune response [46]. In vivo, they found that macrophages from overexpressing ACE mice are more pro-inflammatory and have more antitumor ability compared to those from wild-type mice [47]. These discrepant data emphasized different targets and receptors in RAS system may result in different immune effects of tumors. Thus we need more specific and definite experiments to verify the mechanisms.

Vessel perfusion and hypoxia

The alleviation of solid stress could be associated with the decompression of collapsed vessels, which could improve tumor perfusion and decrease hypoxia. In human ovarian tumor and pancreatic tumor cells that were treated with losartan, an increase in the percentage of perfused vessels and a decrease in hypoxic levels has been reported [2,28]. As a result of the lowered hypoxia conditions, the expression of VEGF was then reduced [48,49]. However, there are conflicting reports on the levels of VEGF, which is inconsistent with the reports that AT1R expression correlates with VEGF and micro-vessel density (MVD) in different human tumors [29,30,50]. Further research is required to settle these discrepancies.

The role of RASi in tumor process

Except the above mentioned supportive mechanisms of RASi played in tumor, a comprehensive review about the relationship between ARB/ACEI and neoplasms on different angles of tumor process (including genesis, treatment and recurrence) was made. In brief, there are three different perspectives on this subject. A majority of the reports hold the view that ARB/ACEI may be a potential drug or therapy when combined with chemo-radiotherapy/targeted therapy to protect against tumor growth, promote the effectiveness of the different cancer therapies and even reduce the recurrence of carcinoma. On the other hand, there is an opposing view that the use of these RASi could actually increase the risk of cancer. A third perspective suggests that there is not an increased risk among those ARB/ACEI users, but in the meanwhile without definitive evidence to confirm the relationship between the two ARB/ACEI and tumors were positive.

The effect of RASi in tumor genesis

There are two conflicting opinions on the use of RASi in reducing cancer incidence. On one hand, some retrospective analyses demonstrated a reduction in colorectal cancer incidence, polyp formation, and distant metastasis in patients taking RASi [51-53]. Others showed that ARB/ACEI just does not have a risk of developing cancer but cannot imply conclusive findings on the positive effect that RASi may have. We can learn this view from a review conducted by The U.S. Food and Drug Administration (FDA) [54]. In this trial-level meta-analysis of clinical trials, researchers divided patients into two groups (ARB or non-ARB treatment) randomly. They included 31 trials and roughly 156,000 patients and did not find an elevated risk of cancer in the users taking any ARB drugs. In addition, a cohort research carried out among United States veterans also supported the conclusions of the FDA [55]. They identified a total of 543,824 unique veterans and classified them into either ARB treated or non-treated in a 1:15 ratio. A tiny but statistically significant reduction was observed in the incidence of clinically detected prostate cancer (PrCA) among patients receiving ARB treatment. On the other hand, a nationwide high-risk cohort study conducted in Taiwan was designed to analyze the comparative effectiveness of any RASi drugs in the chemoprevention of hepatocellular carcinoma (HCC) in high-risk cohorts [56]. It was revealed that compared with other high risk and protective factors for HCC, RASi use in the viral hepatitis type B (HBV) and viral hepatitis type C (HCV)-the two established risk factors for HCC cohorts did not provide sufficient protection when used at standard dosages and also was not completely safe. What is more, a meta-analysis of randomized controlled trials found that the risk of cancer was higher in treatment groups than control group [22]. Last but not least, a population based cohort study included 992,061 patients who were prescribed with antihypertensive drugs from 1 January 1995 to 31 December 2015 and followed until 31 December 2016 and were classified into three mutually exclusive exposure categories: ACEI (335,135 patients), ARB (29,008 patients) and other anti-hypertensive drugs [24]. It was found that the use of ACEI was associated with an increased risk of lung cancer with 7952 lung cancer events occurring. Moreover, the association was more apparent among people using ACEI for more than five years [55].

These discrepant results may give us a hint that clinical trial designs, different tumor types, and follow up duration of patients all could be the impacting factors in delivering the effects of RASi against cancer. To obtain a definitive conclusion, large scale clinical trials with longer term follow up for each type of tumor are urgently required [57].

The therapeutic effect of ARB/ACEI

RASi use alone

Whether the sole use of any ARB/ACEI agents reduce cancer risk remains an issue to debate. Perhaps due to different limitations, such as healthy user bias (the inclusion of prevalent drug users), outcome detection bias or time-related bias, the clinical data is inconclusive [58-60]. Chen et al compared patients using ARB/ACEI drugs with the control group in esophageal squamous cell carcinoma. The median overall survival was 75.3 months for the ARB/ACEI group, and 58.3 months for the control (non-ARB/ACEI) group. In addition, the months of disease-free survival in the treatment group (50 months) was almost twice than that of the control group (28.6 months). The result may be attributed to the growth inhibition of the esophageal squamous cell carcinoma cell and VEGF secretion as reported previously [61]. Nonetheless, a population-based case control study reported a tiny increased risk with ARB/ACEI usage [62]. Meanwhile, one cohort study aiming at estimating the effects of RASi versus other antihypertensive agents on short term colorectal cancer (CRC) risk, concluded that there is no correlation between RASi administration and the short-term CRC risk [63]. But due to its short follow-up duration, the long-term effect needs to be investigated in the future. Apart from the clinical research, basic experimental studies have also provided differing opinions. In terms of tumor weight or growth, the use of losartan showed no significant positive effect and possibly even a worse outcome. Despite multiple reports have proved that losartan is a factor in promoting changes in the TME, the result related to sole use of losartan on ECM is still unclear. Some reports have indicated that the amount of α-SMA and collagen I (the crucial content in ECM) may have been sharply reduced after treatment with losartan [2,28]. But the result in Chun-hua et al suggests that the effect of losartan on α-SMA and collagen I has almost no difference as compared to the control group [64].

RASi usage in conjunction with chemo-radiotherapy

For metastatic colorectal cancer (mCRC), enrolled patients firstly treated with first-line oxaliplatin-based chemotherapy in combination with bevacizumab, were divided into ARB and non-ARB groups. The ARB group showed longer progression-free survival (PFS) and overall survival (OS) [65]. These beneficial effects are not limited to mCRC, but also can been reported in other cancers, such as glioblastoma (GBM), HCC, advanced gastric cancer and advanced pancreatic cancer, to name a few [66-69]. A late and authoritative phase II clinical trial demonstrated that total neoadjuvant therapy and chemo-radiotherapy in combination with losartan was effective in down-staging locally advanced pancreatic cancer and helpful in improving R0 resection rate to 61% [70]. In another neoadjuvant treatment of rectal cancer, Zachary et al found the rate of pathologic complete response (pCR) to neoadjuvant radiation therapy among patients taking ARB/ACEI is two or three times the amount of those not taking an ARB/ACEI in two data sets [71]. Furthermore, compared with RASi, the results also were not found among other drugs, including statins, metformin, and aspirin. One newest completed clinical trial (NCT01821729) targeted pancreatic cancer by combining FOLFIRINOX (oxaliplatin+irinotecan+5fluorouracil), losartan and proton beam radiation treatments. It was found that 73.5% of participants were given a lower score for Eastern Cooperative Oncology Group Performance Status (ECOG PS), which means better functioning of daily living abilities and activities of the patients. Among all these mentioned studies, they both generally attributed the improved outcomes to two different mechanisms: the role RASIs played in the inhibition of TME and/or down-regulation of VEGF expression [69,70,74]. However, these conclusions fail to explain several points, for instance, how the two combined therapies work synergistically or connect each other in different molecular pathways. To work out this could help us determine which kind of RASi drugs combined with different chemotherapies is the best for specific tumors.

Secondly, some preclinical studies have demonstrated that certain RASi might aid the alleviation of the radiation-induced injury of normal tissue without impairing the tumor response to radiation [72-77]. This effect, on one hand, could be due to the inhibition of ARB/AT1R signaling. Radiation has been shown to up-regulate the expression of Ang II, which contributes to the tissue damage and remodeling via the up-regulation of pro-fibrogenic (like the transforming growth factor β (TGF-β) pathway) and pro-inflammatory pathways [78,79]. On the other hand, increased Ang (1-7) expression seems to be protective towards radiotherapy adverse effects [80,81].

RASi usage in conjunction with targeted therapy

Increasing evidence indicates that it is worth exploring the combined use of ARB/ACEI with targeted therapy. Especially when combined with VEGF therapies in metastatic renal cell carcinoma (mRCC) agents. Recently, studies have shown that the concomitant use of RASi might significantly improve OS and PFS in mRCC patients receiving sunitinib [82,83]. McKay et al conducted a retrospective analysis of people with mRCC who underwent targeted therapy [84]. It was found that RASi users had improved OS as compared with other antihypertensive drugs patients and individuals not receiving antihypertensive therapy. More interestingly, the benefit in OS was limited to RASi users receiving VEGF-targeted therapy rather than in patients taking other drugs (temsirolimus or interferon α). Similarly, another mRCC study sought to determine if there is an effective concomitant use of RASi on the outcome of sunitinib [85]. They showed an improved outcome of sunitinib after the use of RASi, providing evidence for the prospective future combinatorial therapy. The reason why the combinatorial use leads to greater effects is related to the independent role of RASi plays in the tumor cell proliferation, VEGF and angiogenesis. Similar to the effect in chemo-radiotherapy, it has been reported that in lung cancer patients receiving bevacizumab, the administration of RASi helps to reduce the risk of proteinuria, which may occur due to the higher dose of bevacizumab [86,87]. However, the exact drug interactions between the RASIs and other targeted drugs still needs to be investigated to elucidate the precise underlying molecular mechanisms.

Taken together, RASi have a clear potential in combination with chemo-radiotherapy or targeted therapy (following Table 1). However, larger studies are needed to better understand the protective effects of long term RASi use after chemotherapy and the specific molecular mechanisms.

Table 1.

Completed and available prospective/retrospective trials of RASi combine with other therapies to fight against cancer

NCT number Classified & Design Interventions Conditions Outcome
NCT01805453 CHT+RT+ARB; RCT Losartan 50 mg*2/day Newly-diagnosed No results posted
Placebo glioblastoma
NCT01434134 CHT+RT+ARB; RCT Metoprolol 100 mg/d Breast cancer No results posted
Placebo Heart failure
Candesartan 32 mg/d
NCT00459771 Targeted therapy+ARB; RCT Candesartan 32 mg/d Breast cancer No results posted
Placebo
NCT00162955 RT+ARB, RCT Valsartan 80 mg/d Non-hodgkin’s Positive
lymphoma
NCT01110824 CHT+ACEI; RCT Enalapril 2.5 to 10 mg/d Acute myeloid leukemia Positive
Carvedilol 6.25 to 25 mg Lymphoid neoplasm
Multiple myeloma
Lymphoma
NCT02651415 Targeted therapy+ACEI; Single arm Regorafenibn 10 mg/d Metastatic Positive
Perindopril 4 mg/d colorectal cancer
NCT00895414 CHT+ACEI; RCT Doxorubicin hydrochloride Breast cancer Positive
Enalapril maleate
NCT01754909 RT+ACEI; RCT Enalapril 2.5, 5, or 10 mg Lung cancer Positive
escalating doses Placebo Radiation pneumonitis

RT: radiation therapy; RCT: randomized control therapy; CHT: chemotherapy.

The effect of RASi in tumor recurrence

In this section, we discuss the beneficial effects that ARB/ACEI may have on the recurrence of tumors. One study has analyzed the medical records of patients diagnosed with stage II/III breast cancer from 1999 to 2005 [88]. A total of 168 patients who used ACEI or ARB for at least 6 months were included in the analysis. Moreover, the most frequent agents they used were lisinopril and valsartan. At the end of the study, 15% of the ARB or ACEI patients recurred, which is less than the rate of non-users (23%). Also, the disease-free survival of ARB/ACEI users, was significantly higher than non-drug users. These highlight that the use of ARB/ACEI was associated with a reduced risk of breast cancer recurrence. Another study also supported that RASi was associated with reduced risk of pathologic N3 (pN3) breast cancer recurrence [89]. A randomized controlled trial (in Japan) was designed to explore the collaborative effect of vitamin K2 (VK) and ACEI (after curative treatment) on cumulative recurrence of HCC [90]. The results revealed that after administrating VK (menatetrenone; 45 mg/day) and/or ACEI (perindopril; 4 mg/day) for 48 months, the combination treatment markedly inhibited the cumulative recurrence of HCC in association with the suppression of VEGF. Nonetheless, when using VK or ACEI alone, these beneficial effects were not observed The idea that a single anti-angiogenic agent may not be sufficient to completely inhibit the tumor angiogenesis might explain this result [91]. The similar benefit is not only observed for breast cancer and HCC but also in the recurrence of the CRC, especially in left-sided CRC and early-stage CRC [92]. However, in one Danish nationwide prospective cohort study, which followed patients for 10 years, researchers found that there was an almost null association between the use of any RASi drugs and breast cancer recurrence, compared with non-users (ACEI: HR: 1.2, 95% CI, 0.97 to 1.4; ARBs: HR: 1.1, 95% CI, 0.85 to 1.3) [93]. In the Kaiser Permanente Northern California Cancer Registry, one study observed women with early stage breast cancer and examined the association between ACEI and breast cancer recurrence, breast cancer-specific mortality, and overall mortality [94]. In their Cox proportional hazards models, the exposure of ACEI was associated with breast cancer recurrence (HR 1.56, 95% CI 1.02, 2.39, P = 0.04), but did not cause specific or overall mortality.

Discussion and prospect

Substantial reviews and meta-analyses have reported this subject since many years ago [95]. For example, the previous reviews had elucidated the potential mechanisms of RASi in tumor angiogenesis, cell proliferation and metastasis. Also, they pointed towards the promising possibilities that ARB/ACEI may have in different types of human cancers [96-99]. Or with systematic literature research, some meta-analysis reached the conclusion whether the exposure of RASi increased the risk of cancer [22,100,101]. Different from these reviews, we focused more on the mechanisms of regulating TME and improving hypoxia by RASi, which are both the keen directions in nowadays cancer treatment. What’s more, we have thoroughly analyzed its effects on chemo-radiotherapy and targeted therapy from cancer genesis to recurrence. Although it is not conclusive that RASi increases or decreases the incidence of cancer, most experiments so far suggest RAS blockade could slow down tumor process from various aspects. More specifically, compared with separate usage, lots of evidence are inclined to use RASi as an add-on to standard cancer treatment (just as the ongoing prospective trials showed in the Table 2). At the end, we put the existing controversial points together, suggesting that the current studies still cannot provide a consistent answer for cancer associated with RASi exposure.

Table 2.

Ongoing prospective trials of RASi combine with other therapies to fight against cancer

NCT number Classified & Design conditions Interventions Status
NCT03563248 RT+CHT+ARB+Targeted therapy; RCT Pancreatic Cancer FOLFIRINOX; Losartan Recruiting
Nivolumab; SBRT; Surgery
NCT03864042 ARB+RT; N-RCT (Sequential Assignment) Advanced Solid Tumors Losartan: 25 mg (day 1, 7, 14) Recruiting
Metastatic Melanoma Caffeine: 50 mg (day 1, 7, 14)
Omeprazole: 20 mg (day 1, 7, 14)
Midazolam: 2 mg (day 1, 7, 14)
Encorafenib: 450 mg/d
Binimetinib: 45 mg/d
NCT03951142 ARB+CHT+TT; RCT Glioblastoma Losartan Recruiting
Brain Metastases
NCT03900793 ARB+TT; N-RCT (Sequential Assignment) Osteosarcoma Losartan: 12.5~100 mg, escalating dose Recruiting
Sunitinib
NCT04092309 ACEI+ARNI; RCT Hematopoietic Stem Cell Transplantation Cardiotoxicity ACEI; Sacubitril-Valsartan Recruiting
NCT03389724 ACEI+CHT; RCT Cardiotoxicity ACEI (Capoten): 0.5 mg/kg/day Recruiting
Acute Myeloid Leukemia in Children
Bone tumor
NCT04288700 ACEI+β-receptor blocker; RCT Infantile hemangioma Oral propranolol: 2 mg/kg/d Recruiting
Oral Captopril: 0.1~0.3 mg/kg/d
intralesional propranolol injection: 1 mg/mL
NCT02770378 ACEI+CHT; Single arm Glioblastoma Temozolomide: 20 mg/m2 BSA *2/d Active, not recruiting
Aprepitant: 80 mg/d
Minocycline: 50 or 100 mg*2/d
Captopril: 25 or 50 mg*2/d
NCT02236806 ACEI+β-receptor blocker; RCT Breast Cancer Bisoprolol: 5 mg/d Recruiting
Cardiotoxicit Ramipril: 5 mg/d
Placebo: 1 capsule/d
NCT03475186 ACEI+RRT; Single arm Glioblastoma Ramipril: 2.5~5/d Recruiting
Radiotherapy; Complications
Cognitive Decline
Chemoradiation
NCT01646437 ACEI+β-receptor blocker+thiazide; RCT Cardiovascular Disease Hydrochlorothiazide: 25 mg/d; Atenolol: 100 mg/d, Ramipril: 10 mg/d; Simvastatin: 40 mg/d; Placebo Active, not recruiting
Fractures
Cancers
NCT02596126 ACEI+aspirin; RCT Myocardial Infarction Aspirin: 100 mg/d, Atorvastatin 40 mg/d; Ramipril: 2.5 mg, or 5 mg, or 10 mg/d Recruiting
Cardiovascular Disease
NCT01968200 ACEI; RCT Cancer Enalapril: 2.5~10 mg/12 h Active, not recruiting

RT: radiation therapy; RCT: randomized control therapy; CHT: chemotherapy.

The reason why there are still some inconsistent results is that different cancer types and the exact receptors of RASi are both the causes which affect the anti-cancer effects. Like AT1R has contradictory effects in different tumors. In breast and pancreatic cancers, AT1R positively regulates tumor growth but it negatively regulates growth in melanoma cancer [2,31]. The different expression of Ang II receptors among the different types of cancer and patients may be another reason [95,102]. It has demonstrated that peroxisome proliferator-activated receptor gamma (PPARγ) impacts tumor progression by affecting cell proliferation, apoptosis and angiogenesis in TME [103]. Losartan and telmisartan are demonstrated as partial PPARγ activators that impede cell proliferation through inducing cell cycle arrest [104,105]. Whether different types of ARBs are the factors that could lead to different extent of PPARγ activation is yet largely unclear [57]. Though the multimodal effects of ACEIs/ARBs in cancer angiogenesis and TME provide us insights about cancer treatment and outcome, we are just at the beginning to elucidate the precise and sophisticated mechanism of RAS in various cancers.

Further research is required to explore dose and duration of RASi in different cancer types and identify the predictive or prognostic biomarkers as well. Generally speaking, firstly for patient diagnosed with hypertension and cancer, the use of RASi could be a promising strategy. But for a patient at normal tension, exploring an optimal dose and administration ways to avoid hypotension and other side effects is one problem that must be overcame during the use of RASi. Secondly perhaps biomaterials-assisted local treatment strategies are able to enhance anticancer outcomes and reduce systematic toxicity. For example, Chauhan et al has successfully designed a nano-conjugates to preferentially accumulate and act in tumors and consequently enhanced anti-tumor effects [38]. Thirdly studies have pointed out that normalized tumor vasculature in TME impacts both the tumor progression and the efficacy of all types of anti-tumor therapies [15]. Based on their advances, except the above combined therapies, RASis may be also combined with immune checkpoint blockers in immune-modulatory to achieve better anti-tumor effects. Lastly, other than just focusing on inhibiting tumors (such as pancreatic cancer, breast cancer, GBM), the role that RASi plays in mitigating cancer treatment-related adverse events (especially cardio toxicity) should also be priority. To decipher and make sense this point, larger and more credible verifications are required.

Conclusions

In general, there are lots of compelling clinical and basic reports that support the role of ARB/ACEI drugs in tumor treatment with its role in angiogenesis, ECM, TME and hypoxia. Of all the established therapies, using it as an enhancement to chemo-radiotherapy or targeted therapy is prospectively beneficial to fight against tumor. However, we still cannot ignore reports that show the opposite or conflicting views. Thus, larger and persuasive studies are indispensable to clarify this subject.

Disclosure of conflict of interest

None.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7–30. doi: 10.3322/caac.21590. [DOI] [PubMed] [Google Scholar]
  • 2.Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV, Stylianopoulos T, Mousa AS, Han X, Adstamongkonkul P, Popović Z, Huang P, Bawendi MG, Boucher Y, Jain RK. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4:2516. doi: 10.1038/ncomms3516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lee ST, Muralidharan V, Tebbutt N, Wong P, Fang C, Liu Z, Gan H, Sachinidis J, Pathmaraj K, Christophi C, Scott AM. Prevalence of hypoxia and correlation with glycolytic metabolism and angiogenic biomarkers in metastatic colorectal carcinoma. Eur J Nucl Med Mol Imaging. 2020 doi: 10.1007/s00259-020-05074-5. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 4.Katsi V, Zerdes I, Manolakou S, Makris T, Nihoyannopoulos P, Tousoulis D, Kallikazaros I. Anti-VEGF anticancer drugs: mind the hypertension. Recent Adv Cardiovasc Drug Discov. 2014;9:63–72. doi: 10.2174/1574890110999150604114127. [DOI] [PubMed] [Google Scholar]
  • 5.Moslehi JJ, Deininger M. Tyrosine kinase inhibitor-associated cardiovascular toxicity in chronic myeloid leukemia. J. Clin. Oncol. 2015;33:4210–4218. doi: 10.1200/JCO.2015.62.4718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jain RK. Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J. Clin. Oncol. 2013;31:2205–2218. doi: 10.1200/JCO.2012.46.3653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sorensen AG, Emblem KE, Polaskova P, Jennings D, Kim H, Ancukiewicz M, Wang M, Wen PY, Ivy P, Batchelor TT, Jain RK. Increased survival of glioblastoma patients who respond to antiangiogenic therapy with elevated blood perfusion. Cancer Res. 2012;72:402–407. doi: 10.1158/0008-5472.CAN-11-2464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Park MS, Klotz E, Kim MJ, Song SY, Park SW, Cha SW, Lim JS, Seong J, Chung JB, Kim KW. Perfusion CT: noninvasive surrogate marker for stratification of pancreatic cancer response to concurrent chemo- and radiation therapy. Radiology. 2009;250:110–117. doi: 10.1148/radiol.2493080226. [DOI] [PubMed] [Google Scholar]
  • 9.Pinter M, Jain RK. Targeting the renin-angiotensin system to improve cancer treatment: implications for immunotherapy. Sci Transl Med. 2017;9:eaan5616. doi: 10.1126/scitranslmed.aan5616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats AJ, Falk V, Gonzalez-Juanatey JR, Harjola VP, Jankowska EA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B, Riley JP, Rosano GM, Ruilope LM, Ruschitzka F, Rutten FH, van der Meer P Authors/Task Force Members; Document Reviewers. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail. 2016;18:891–975. doi: 10.1002/ejhf.592. [DOI] [PubMed] [Google Scholar]
  • 11.Levin A, Stevens PE. Summary of KDIGO 2012 CKD guideline: behind the scenes, need for guidance, and a framework for moving forward. Kidney Int. 2014;85:49–61. doi: 10.1038/ki.2013.444. [DOI] [PubMed] [Google Scholar]
  • 12.James PA, Oparil S, Carter BL, Cushman WC, Dennison-Himmelfarb C, Handler J, Lackland DT, LeFevre ML, MacKenzie TD, Ogedegbe O, Smith SC Jr, Svetkey LP, Taler SJ, Townsend RR, Wright JT Jr, Narva AS, Ortiz E. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8) JAMA. 2014;311:507–520. doi: 10.1001/jama.2013.284427. [DOI] [PubMed] [Google Scholar]
  • 13.O’Gara PT, Kushner FG, Ascheim DD, Casey DE Jr, Chung MK, de Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, Granger CB, Krumholz HM, Linderbaum JA, Morrow DA, Newby LK, Ornato JP, Ou N, Radford MJ, Tamis-Holland JE, Tommaso CL, Tracy CM, Woo YJ, Zhao DX. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;61:485–510. doi: 10.1016/j.jacc.2012.11.018. [DOI] [PubMed] [Google Scholar]
  • 14.Cronin-Fenton D. Angiotensin converting enzyme inhibitors and lung cancer. BMJ. 2018;363:k4337. doi: 10.1136/bmj.k4337. [DOI] [PubMed] [Google Scholar]
  • 15.Perini MV, Dmello RS, Nero TL, Chand AL. Evaluating the benefits of renin-angiotensin system inhibitors as cancer treatments. Pharmacol Ther. 2020;211:107527. doi: 10.1016/j.pharmthera.2020.107527. [DOI] [PubMed] [Google Scholar]
  • 16.Sun H, Li T, Zhuang R, Cai W, Zheng Y. Do renin-angiotensin system inhibitors influence the recurrence, metastasis, and survival in cancer patients?: evidence from a meta-analysis including 55 studies. Medicine (Baltimore) 2017;96:e6394. doi: 10.1097/MD.0000000000006394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.George AJ, Thomas WG, Hannan RD. The renin-angiotensin system and cancer: old dog, new tricks. Nat Rev Cancer. 2010;10:745–759. doi: 10.1038/nrc2945. [DOI] [PubMed] [Google Scholar]
  • 18.Bader M. Tissue renin-angiotensin-aldosterone systems: targets for pharmacological therapy. Annu Rev Pharmacol Toxicol. 2010;50:439–465. doi: 10.1146/annurev.pharmtox.010909.105610. [DOI] [PubMed] [Google Scholar]
  • 19.Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci U S A. 2011;108:2909–2914. doi: 10.1073/pnas.1018892108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rhodes DR, Ateeq B, Cao Q, Tomlins SA, Mehra R, Laxman B, Kalyana-Sundaram S, Lonigro RJ, Helgeson BE, Bhojani MS, Rehemtulla A, Kleer CG, Hayes DF, Lucas PC, Varambally S, Chinnaiyan AM. AGTR1 overexpression defines a subset of breast cancer and confers sensitivity to losartan, an AGTR1 antagonist. Proc Natl Acad Sci U S A. 2009;106:10284–10289. doi: 10.1073/pnas.0900351106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Neo JH, Malcontenti-Wilson C, Muralidharan V, Christophi C. Effect of ACE inhibitors and angiotensin II receptor antagonists in a mouse model of colorectal cancer liver metastases. J Gastroenterol Hepatol. 2007;22:577–584. doi: 10.1111/j.1440-1746.2006.04797.x. [DOI] [PubMed] [Google Scholar]
  • 22.Sipahi I, Debanne SM, Rowland DY, Simon DI, Fang JC. Angiotensin-receptor blockade and risk of cancer: meta-analysis of randomised controlled trials. Lancet Oncol. 2010;11:627–636. doi: 10.1016/S1470-2045(10)70106-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hicks BM, Filion KB, Yin H, Sakr L, Udell JA, Azoulay L. Angiotensin converting enzyme inhibitors and risk of lung cancer: population based cohort study. BMJ. 2018;363:k4209. doi: 10.1136/bmj.k4209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Costache MI, Ioana M, Iordache S, Ene D, Costache CA, Săftoiu A. VEGF expression in pancreatic cancer and other malignancies: a review of the literature. Rom J Intern Med. 2015;53:199–208. doi: 10.1515/rjim-2015-0027. [DOI] [PubMed] [Google Scholar]
  • 25.Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: beyond discovery and development. Cell. 2019;176:1248–1264. doi: 10.1016/j.cell.2019.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ferrara N. Vascular endothelial growth factor and age-related macular degeneration: from basic science to therapy. Nat Med. 2010;16:1107–1111. doi: 10.1038/nm1010-1107. [DOI] [PubMed] [Google Scholar]
  • 27.Zhao B, Sun H, Zhang XD, Li GH, Zhao YH, Wang B. Potential anti-tumor mechanisms of renin angiotensin system inhibitors through inhibiting angiogenesis and influencing angiotensin II actions. Int J Clin Exp Med. 2018;11:10585–10594. [Google Scholar]
  • 28.Zhao Y, Cao J, Melamed A, Worley M, Gockley A, Jones D, Nia HT, Zhang Y, Stylianopoulos T, Kumar AS, Mpekris F, Datta M, Sun Y, Wu L, Gao X, Yeku O, Del Carmen MG, Spriggs DR, Jain RK, Xu L. Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma. Proc Natl Acad Sci U S A. 2019;116:2210–2219. doi: 10.1073/pnas.1818357116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Arrieta O, Villarreal-Garza C, Vizcaíno G, Pineda B, Hernández-Pedro N, Guevara-Salazar P, Wegman-Ostrosky T, Villanueva-Rodríguez G, Gamboa-Domínguez A. Association between AT1 and AT2 angiotensin II receptor expression with cell proliferation and angiogenesis in operable breast cancer. Tumour Biol. 2015;36:5627–5634. doi: 10.1007/s13277-015-3235-3. [DOI] [PubMed] [Google Scholar]
  • 30.Ino K, Shibata K, Kajiyama H, Yamamoto E, Nagasaka T, Nawa A, Nomura S, Kikkawa F. Angiotensin II type 1 receptor expression in ovarian cancer and its correlation with tumour angiogenesis and patient survival. Br J Cancer. 2006;94:552–560. doi: 10.1038/sj.bjc.6602961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Renziehausen A, Wang H, Rao B, Weir L, Nigro CL, Lattanzio L, Merlano M, Vega-Rioja A, Del Carmen Fernandez-Carranco M, Hajji N, Matin R, Harwood C, Li S, Sim VR, O’Neill K, Evans A, Thompson A, Szlosarek P, Fleming C, Stebbing J, Proby C, Tzakos AG, Syed N, Crook T. The renin angiotensin system (RAS) mediates bifunctional growth regulation in melanoma and is a novel target for therapeutic intervention. Oncogene. 2019;38:2320–2336. doi: 10.1038/s41388-018-0563-y. [DOI] [PubMed] [Google Scholar]
  • 32.Azevedo H, Fujita A, Bando SY, Iamashita P, Moreira-Filho CA. Transcriptional network analysis reveals that AT1 and AT2 angiotensin II receptors are both involved in the regulation of genes essential for glioma progression. PLoS One. 2014;9:e110934. doi: 10.1371/journal.pone.0110934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nguyen L, Ager EI, Neo J, Christophi C. Regulation of colorectal cancer cell epithelial to mesenchymal transition by the renin angiotensin system. J Gastroenterol Hepatol. 2016;31:1773–1782. doi: 10.1111/jgh.13307. [DOI] [PubMed] [Google Scholar]
  • 34.Chen XM, Song EW. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discovery. 2019;18:99–115. doi: 10.1038/s41573-018-0004-1. [DOI] [PubMed] [Google Scholar]
  • 35.Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028–1040. doi: 10.1038/nm.2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401. doi: 10.1038/nrc1877. [DOI] [PubMed] [Google Scholar]
  • 37.Piper M, Mueller AC, Karam SD. The interplay between cancer associated fibroblasts and immune cells in the context of radiation therapy. Mol Carcinog. 2020;59:754–765. doi: 10.1002/mc.23205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chauhan VP, Chen IX, Tong R, Ng MR, Martin JD, Naxerova K, Wu MW, Huang P, Boucher Y, Kohane DS, Langer R, Jain RK. Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy. Proc Natl Acad Sci U S A. 2019;116:10674–10680. doi: 10.1073/pnas.1819889116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Helmlinger G, Netti PA, Lichtenbeld HC, Melder RJ, Jain RK. Solid stress inhibits the growth of multicellular tumor spheroids. Nat Biotechnol. 1997;15:778–783. doi: 10.1038/nbt0897-778. [DOI] [PubMed] [Google Scholar]
  • 40.Nia HT, Datta M, Seano G, Huang P, Munn LL, Jain RK. Quantifying solid stress and elastic energy from excised or in situ tumors. Nat Protoc. 2018;13:1091–1105. doi: 10.1038/nprot.2018.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tone A, Shikata K, Ogawa D, Sasaki S, Nagase R, Sasaki M, Yozai K, Usui HK, Okada S, Wada J, Shikata Y, Makino H. Changes of gene expression profiles in macrophages stimulated by angiotensin II--angiotensin II induces MCP-2 through AT1-receptor. J Renin Angiotensin Aldosterone Syst. 2007;8:45–50. doi: 10.3317/jraas.2007.007. [DOI] [PubMed] [Google Scholar]
  • 42.Kosugi M, Miyajima A, Kikuchi E, Horiguchi Y, Murai M. Angiotensin II type 1 receptor antagonist candesartan as an angiogenic inhibitor in a xenograft model of bladder cancer. Clin Cancer Res. 2006;12:2888–2893. doi: 10.1158/1078-0432.CCR-05-2213. [DOI] [PubMed] [Google Scholar]
  • 43.Egami K, Murohara T, Shimada T, Sasaki K, Shintani S, Sugaya T, Ishii M, Akagi T, Ikeda H, Matsuishi T, Imaizumi T. Role of host angiotensin II type 1 receptor in tumor angiogenesis and growth. J Clin Invest. 2003;112:67–75. doi: 10.1172/JCI16645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shirotake S, Miyajima A, Kosaka T, Tanaka N, Kikuchi E, Mikami S, Okada Y, Oya M. Regulation of monocyte chemoattractant protein-1 through angiotensin II type 1 receptor in prostate cancer. Am J Pathol. 2012;180:1008–1016. doi: 10.1016/j.ajpath.2011.11.027. [DOI] [PubMed] [Google Scholar]
  • 45.Shen XZ, Lukacher AE, Billet S, Williams IR, Bernstein KE. Expression of angiotensin-converting enzyme changes major histocompatibility complex class I peptide presentation by modifying C termini of peptide precursors. J Biol Chem. 2008;283:9957–9965. doi: 10.1074/jbc.M709574200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008;222:162–179. doi: 10.1111/j.1600-065X.2008.00602.x. [DOI] [PubMed] [Google Scholar]
  • 47.Shen XZ, Okwan-Duodu D, Blackwell WL, Ong FS, Janjulia T, Bernstein EA, Fuchs S, Alkan S, Bernstein KE. Myeloid expression of angiotensin-converting enzyme facilitates myeloid maturation and inhibits the development of myeloid-derived suppressor cells. Lab Invest. 2014;94:536–544. doi: 10.1038/labinvest.2014.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jin F, Zheng X, Yang Y, Yao G, Ye L, Doeppner TR, Hermann DM, Wang H, Dai Y. Impairment of hypoxia-induced angiogenesis by LDL involves a HIF-centered signaling network linking inflammatory TNFα and angiogenic VEGF. Aging (Albany NY) 2019;11:328–349. doi: 10.18632/aging.101726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nicolas S, Abdellatef S, Haddad MA, Fakhoury I, El-Sibai M. Hypoxia and EGF stimulation regulate VEGF expression in human glioblastoma multiforme (GBM) Cells by differential regulation of the PI3K/Rho-GTPase and MAPK pathways. Cells. 2019;8:1397. doi: 10.3390/cells8111397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shirotake S, Miyajima A, Kosaka T, Tanaka N, Maeda T, Kikuchi E, Oya M. Angiotensin II type 1 receptor expression and microvessel density in human bladder cancer. Urology. 2011;77:7. doi: 10.1016/j.urology.2010.11.002. [DOI] [PubMed] [Google Scholar]
  • 51.Childers WK. Interactions of the renin-angiotensin system in colorectal cancer and metastasis. Int J Colorectal Dis. 2015;30:749–752. doi: 10.1007/s00384-014-2118-1. [DOI] [PubMed] [Google Scholar]
  • 52.Fujita M, Hayashi I, Yamashina S, Itoman M, Majima M. Blockade of angiotensin AT1a receptor signaling reduces tumor growth, angiogenesis, and metastasis. Biochem Biophys Res Commun. 2002;294:441–447. doi: 10.1016/S0006-291X(02)00496-5. [DOI] [PubMed] [Google Scholar]
  • 53.Folkman J. Anti-angiogenesis: new concept for therapy of solid tumors. Ann Surg. 1972;175:409–416. doi: 10.1097/00000658-197203000-00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.U.S. Food and Drug Administration, 2011. No increase in risk of cancer with certain blood pressure drugs - Angiotensin Receptor Blockers (ARBs) [online] Available: http://www.fda.gov/Drugs/DrugSafety/ucm257516.htm [Accessed 22 December 2011]
  • 55.Rao GA, Mann JR, Bottai M, Uemura H, Burch JB, Bennett CL, Haddock KS, Hebert JR. Angiotensin receptor blockers and risk of prostate cancer among United States veterans. J Clin Pharmacol. 2013;53:773–778. doi: 10.1002/jcph.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ho CM, Lee CH, Lee MC, Zhang JF, Wang JY, Hu RH, Lee PH. Comparative effectiveness of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers in chemoprevention of hepatocellular carcinoma: a nationwide high-risk cohort study. BMC Cancer. 2018;18:401. doi: 10.1186/s12885-018-4292-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ishikane S, Takahashi-Yanaga F. The role of angiotensin II in cancer metastasis: Potential of renin-angiotensin system blockade as a treatment for cancer metastasis. Biochem Pharmacol. 2018;151:96–103. doi: 10.1016/j.bcp.2018.03.008. [DOI] [PubMed] [Google Scholar]
  • 58.Shrank WH, Patrick AR, Brookhart MA. Healthy user and related biases in observational studies of preventive interventions: a primer for physicians. J Gen Intern Med. 2011;26:546–550. doi: 10.1007/s11606-010-1609-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.D’Arcy M, Stürmer T, Lund JL. The importance and implications of comparator selection in pharmacoepidemiologic research. Curr Epidemiol Rep. 2018;5:272–283. doi: 10.1007/s40471-018-0155-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Suissa S. Immortal time bias in pharmaco-epidemiology. Am J Epidemiol. 2008;167:492–499. doi: 10.1093/aje/kwm324. [DOI] [PubMed] [Google Scholar]
  • 61.Chen YH, Huang CH, Lu HI, Chen CH, Huang WT, Hsieh MJ, Rau KM, Chang AY, Lin WC, Li SH. Prognostic impact of renin-angiotensin system blockade in esophageal squamous cell carcinoma. J Renin Angiotensin Aldosterone Syst. 2015;16:1185–1192. doi: 10.1177/1470320314535275. [DOI] [PubMed] [Google Scholar]
  • 62.Hallas J, Christensen R, Andersen M, Friis S, Bjerrum L. Long term use of drugs affecting the renin-angiotensin system and the risk of cancer: a population-based case-control study. Br J Clin Pharmacol. 2012;74:180–188. doi: 10.1111/j.1365-2125.2012.04170.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Htoo PT, Sturmer T, Jonsson-Funk M, Pate V, Simpson RJ Jr, Lund JL. Renin-angiotensin-aldosterone system-based antihypertensive agents and the risk of colorectal cancer among medicare beneficiaries. Epidemiology. 2019;30:867–875. doi: 10.1097/EDE.0000000000001065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hu C, Liu X, Ran W, Meng J, Zhai Y, Zhang P, Yin Q, Yu H, Zhang Z, Li Y. Regulating cancer associated fibroblasts with losartan-loaded injectable peptide hydrogel to potentiate chemotherapy in inhibiting growth and lung metastasis of triple negative breast cancer. Biomaterials. 2017;144:60–72. doi: 10.1016/j.biomaterials.2017.08.009. [DOI] [PubMed] [Google Scholar]
  • 65.Osumi H, Matsusaka S, Wakatsuki T, Suenaga M, Shinozaki E, Mizunuma N. Angiotensin II type-1 receptor blockers enhance the effects of bevacizumab-based chemotherapy in metastatic colorectal cancer patients. Mol Clin Oncol. 2015;3:1295–1300. doi: 10.3892/mco.2015.630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kim ST, Park KH, Oh SC, Seo JH, Kim JS, Shin SW, Kim YH. How does inhibition of the renin-angiotensin system affect the prognosis of advanced gastric cancer patients receiving platinum-based chemotherapy? Oncology. 2012;83:354–360. doi: 10.1159/000337979. [DOI] [PubMed] [Google Scholar]
  • 67.Pinter M, Weinmann A, Wörns MA, Hucke F, Bota S, Marquardt JU, Duda DG, Jain RK, Galle PR, Trauner M, Peck-Radosavljevic M, Sieghart W. Use of inhibitors of the renin-angiotensin system is associated with longer survival in patients with hepatocellular carcinoma. United European Gastroenterol J. 2017;5:987–996. doi: 10.1177/2050640617695698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Levin VA, Chan J, Datta M, Yee JL, Jain RK. Effect of angiotensin system inhibitors on survival in newly diagnosed glioma patients and recurrent glioblastoma patients receiving chemotherapy and/or bevacizumab. J Neurooncol. 2017;134:325–330. doi: 10.1007/s11060-017-2528-3. [DOI] [PubMed] [Google Scholar]
  • 69.Nakai Y, Isayama H, Ijichi H, Sasaki T, Sasahira N, Hirano K, Kogure H, Kawakubo K, Yagioka H, Yashima Y, Mizuno S, Yamamoto K, Arizumi T, Togawa O, Matsubara S, Tsujino T, Tateishi K, Tada M, Omata M, Koike K. Inhibition of renin-angiotensin system affects prognosis of advanced pancreatic cancer receiving gemcitabine. Br J Cancer. 2010;103:1644–1648. doi: 10.1038/sj.bjc.6605955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Murphy JE, Wo JY, Ryan DP, Clark JW, Jiang W, Yeap BY, Drapek LC, Ly L, Baglini CV, Blaszkowsky LS, Ferrone CR, Parikh AR, Weekes CD, Nipp RD, Kwak EL, Allen JN, Corcoran RB, Ting DT, Faris JE, Zhu AX, Goyal L, Berger DL, Qadan M, Lillemoe KD, Talele N, Jain RK, DeLaney TF, Duda DG, Boucher Y, Fernández-Del Castillo C, Hong TS. Total neoadjuvant therapy with folfirinox in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer: a phase 2 clinical trial. JAMA Oncol. 2019;5:1020–1027. doi: 10.1001/jamaoncol.2019.0892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Morris ZS, Saha S, Magnuson WJ, Morris BA, Borkenhagen JF, Ching A, Hirose G, McMurry V, Francis DM, Harari PM, Chappell R, Tsuji S, Ritter MA. Increased tumor response to neoadjuvant therapy among rectal cancer patients taking angiotensin-converting enzyme inhibitors or angiotensin receptor blockers. Cancer. 2016;122:2487–2495. doi: 10.1002/cncr.30079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jaggi JS, Seshan SV, McDevitt MR, Sgouros G, Hyjek E, Scheinberg DA. Mitigation of radiation nephropathy after internal alpha-particle irradiation of kidneys. Int J Radiat Oncol Biol Phys. 2006;64:1503–1512. doi: 10.1016/j.ijrobp.2005.11.036. [DOI] [PubMed] [Google Scholar]
  • 73.Mahmood J, Jelveh S, Zaidi A, Doctrow SR, Medhora M, Hill RP. Targeting the Renin-angiotensin system combined with an antioxidant is highly effective in mitigating radiation-induced lung damage. Int J Radiat Oncol Biol Phys. 2014;89:722–728. doi: 10.1016/j.ijrobp.2014.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Robbins ME, Zhao W, Garcia-Espinosa MA, Diz DI. Renin-angiotensin system blockers and modulation of radiation-induced brain injury. Curr Drug Targets. 2010;11:1413–1422. doi: 10.2174/1389450111009011413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stewart FA, Akleyev AV, Hauer-Jensen M, Hendry JH, Kleiman NJ, Macvittie TJ, Aleman BM, Edgar AB, Mabuchi K, Muirhead CR, Shore RE, Wallace WH. ICRP publication 118: ICRP statement on tissue reactions and early and late effects of radiation in normal tissues and organs--threshold doses for tissue reactions in a radiation protection context. Ann ICRP. 2012;41:1–322. doi: 10.1016/j.icrp.2012.02.001. [DOI] [PubMed] [Google Scholar]
  • 76.Kohl RR, Kolozsvary A, Brown SL, Zhu G, Kim JH. Differential radiation effect in tumor and normal tissue after treatment with ramipril, an angiotensin-converting enzyme inhibitor. Radiat Res. 2007;168:440–445. doi: 10.1667/RR0707.1. [DOI] [PubMed] [Google Scholar]
  • 77.Bracci S, Valeriani M, Agolli L, De Sanctis V, Maurizi Enrici R, Osti MF. Renin-angiotensin system inhibitors might help to reduce the development of symptomatic radiation pneumonitis after stereotactic body radiotherapy for lung cancer. Clin Lung Cancer. 2016;17:189–197. doi: 10.1016/j.cllc.2015.08.007. [DOI] [PubMed] [Google Scholar]
  • 78.Cao S, Wu R. Expression of angiotensin II and aldosterone in radiation-induced lung injury. Cancer Biol Med. 2012;9:254–260. doi: 10.7497/j.issn.2095-3941.2012.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Robbins ME, Diz DI. Pathogenic role of the renin-angiotensin system in modulating radiation-induced late effects. Int J Radiat Oncol Biol Phys. 2006;64:6–12. doi: 10.1016/j.ijrobp.2005.08.033. [DOI] [PubMed] [Google Scholar]
  • 80.Willey JS, Bracey DN, Gallagher PE, Tallant EA, Wiggins WF, Callahan MF, Smith TL, Emory CL. Angiotensin-(1-7) attenuates skeletal muscle fibrosis and stiffening in a mouse model of extremity sarcoma radiation therapy. J Bone Joint Surg Am. 2016;98:48–55. doi: 10.2106/JBJS.O.00545. [DOI] [PubMed] [Google Scholar]
  • 81.Moore ED, Kooshki M, Metheny-Barlow LJ, Gallagher PE, Robbins ME. Angiotensin-(1-7) prevents radiation-induced inflammation in rat primary astrocytes through regulation of MAP kinase signaling. Free Radic Biol Med. 2013;65:1060–1068. doi: 10.1016/j.freeradbiomed.2013.08.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Izzedine H, Derosa L, Le Teuff G, Albiges L, Escudier B. Hypertension and angiotensin system inhibitors: impact on outcome in sunitinib-treated patients for metastatic renal cell carcinoma. Ann Oncol. 2015;26:1128–1133. doi: 10.1093/annonc/mdv147. [DOI] [PubMed] [Google Scholar]
  • 83.Sorich MJ, Kichenadasse G, Rowland A, Woodman RJ, Mangoni AA. Angiotensin system inhibitors and survival in patients with metastatic renal cell carcinoma treated with VEGF-targeted therapy: a pooled secondary analysis of clinical trials. Int J Cancer. 2016;138:2293–2299. doi: 10.1002/ijc.29972. [DOI] [PubMed] [Google Scholar]
  • 84.McKay RR, Rodriguez GE, Lin X, Kaymakcalan MD, Hamnvik OP, Sabbisetti VS, Bhatt RS, Simantov R, Choueiri TK. Angiotensin system inhibitors and survival outcomes in patients with metastatic renal cell carcinoma. Clin Cancer Res. 2015;21:2471–2479. doi: 10.1158/1078-0432.CCR-14-2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Keizman D, Huang P, Eisenberger MA, Pili R, Kim JJ, Antonarakis ES, Hammers H, Carducci MA. Angiotensin system inhibitors and outcome of sunitinib treatment in patients with metastatic renal cell carcinoma: a retrospective examination. Eur J Cancer. 2011;47:1955–1961. doi: 10.1016/j.ejca.2011.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nihei S, Sato J, Harada T, Kuyama S, Suzuki T, Waga N, Saito Y, Kisara S, Yokota A, Okada K, Tsuchiya M, Terui K, Tadokoro Y, Chiba T, Kudo K, Oizumi S, Inoue A, Morikawa N. Antiproteinuric effects of renin-angiotensin inhibitors in lung cancer patients receiving bevacizumab. Cancer Chemother Pharmacol. 2018;81:1051–1059. doi: 10.1007/s00280-018-3580-1. [DOI] [PubMed] [Google Scholar]
  • 87.Wu S, Kim C, Baer L, Zhu X. Bevacizumab increases risk for severe proteinuria in cancer patients. J Am Soc Nephrol. 2010;21:1381–1389. doi: 10.1681/ASN.2010020167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chae YK, Valsecchi ME, Kim J, Bianchi AL, Khemasuwan D, Desai A, Tester W. Reduced risk of breast cancer recurrence in patients using ACE inhibitors, ARBs, and/or statins. Cancer Invest. 2011;29:585–593. doi: 10.3109/07357907.2011.616252. [DOI] [PubMed] [Google Scholar]
  • 89.Babacan T, Balakan O, Kuzan TY, Sarici F, Koca E, Kertmen N, Petekkaya I, Altundag K. The effect of renin-angiotensin-system inhibition on survival and recurrence of N3+breast cancer patients. J BUON. 2015;20:50–56. [PubMed] [Google Scholar]
  • 90.Yoshiji H, Noguchi R, Toyohara M, Ikenaka Y, Kitade M, Kaji K, Yamazaki M, Yamao J, Mitoro A, Sawai M, Yoshida M, Fujimoto M, Tsujimoto T, Kawaratani H, Uemura M, Fukui H. Combination of vitamin K2 and angiotensin-converting enzyme inhibitor ameliorates cumulative recurrence of hepatocellular carcinoma. J Hepatol. 2009;51:315–321. doi: 10.1016/j.jhep.2009.04.011. [DOI] [PubMed] [Google Scholar]
  • 91.Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer. 2004;4:423–436. doi: 10.1038/nrc1369. [DOI] [PubMed] [Google Scholar]
  • 92.Ozawa T, Hashiguchi Y, Yagi T, Fukushima Y, Shimada R, Hayama T, Tsuchiya T, Nozawa K, Iinuma H, Ishihara S, Matsuda K. Angiotensin I-converting enzyme inhibitors/angiotensin II receptor blockers may reduce tumor recurrence in left-sided and early colorectal cancers. Int J Colorectal Dis. 2019;34:1731–1739. doi: 10.1007/s00384-019-03379-y. [DOI] [PubMed] [Google Scholar]
  • 93.Sørensen GV, Ganz PA, Cole SW, Pedersen LA, Sørensen HT, Cronin-Fenton DP, Garne JP, Christiansen PM, Lash TL, Ahern TP. Use of β-blockers, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, and risk of breast cancer recurrence: a Danish nationwide prospective cohort study. J. Clin. Oncol. 2013;31:2265–2272. doi: 10.1200/JCO.2012.43.9190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ganz PA, Habel LA, Weltzien EK, Caan BJ, Cole SW. Examining the influence of beta blockers and ACE inhibitors on the risk for breast cancer recurrence: results from the LACE cohort. Breast Cancer Res Treat. 2011;129:549–556. doi: 10.1007/s10549-011-1505-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Deshayes F, Nahmias C. Angiotensin receptors: a new role in cancer? Trends Endocrinol Metab. 2005;16:293–299. doi: 10.1016/j.tem.2005.07.009. [DOI] [PubMed] [Google Scholar]
  • 96.Miyajima A, Kikuchi E, Kosaka T, Oya M. Angiotensin II type 1 receptor antagonist as an angiogenic inhibitor in urogenital cancer. Rev Recent Clin Trials. 2009;4:75–78. doi: 10.2174/157488709788185996. [DOI] [PubMed] [Google Scholar]
  • 97.Rosenthal T, Gavras I. Angiotensin inhibition and malignancies: a review. J Hum Hypertens. 2009;23:623–635. doi: 10.1038/jhh.2009.21. [DOI] [PubMed] [Google Scholar]
  • 98.Rosenthal T, Gavras I. Renin-angiotensin inhibition in combating malignancy: a review. Anticancer Res. 2019;39:4597–4602. doi: 10.21873/anticanres.13639. [DOI] [PubMed] [Google Scholar]
  • 99.Olin JL, Veverka A, Nuzum DS. Risk of cancer associated with the use of angiotensin II-receptor blockers. Am J Health Syst Pharm. 2011;68:2139–2146. doi: 10.2146/ajhp100570. [DOI] [PubMed] [Google Scholar]
  • 100.Datzmann T, Fuchs S, Andree D, Hohenstein B, Schmitt J, Schindler C. Systematic review and meta-analysis of randomised controlled clinical trial evidence refutes relationship between pharmacotherapy with angiotensin-receptor blockers and an increased risk of cancer. Eur J Intern Med. 2019;64:1–9. doi: 10.1016/j.ejim.2019.04.019. [DOI] [PubMed] [Google Scholar]
  • 101.Hu X, Chen J. Association of angiotensin II type 1 receptor gene A1166C polymorphism with cancer risk: an updated meta-analysis. J Renin Angiotensin Aldosterone Syst. 2019;20:1470320319827207. doi: 10.1177/1470320319827207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Röcken C, Röhl FW, Diebler E, Lendeckel U, Pross M, Carl-McGrath S, Ebert MP. The angiotensin II/angiotensin II receptor system correlates with nodal spread in intestinal type gastric cancer. Cancer Epidemiol Biomarkers Prev. 2007;16:1206–1212. doi: 10.1158/1055-9965.EPI-05-0934. [DOI] [PubMed] [Google Scholar]
  • 103.Reka AK, Goswami MT, Krishnapuram R, Standiford TJ, Keshamouni VG. Molecular cross-regulation between PPAR-γ and other signaling pathways: implications for lung cancer therapy. Lung Cancer. 2011;72:154–159. doi: 10.1016/j.lungcan.2011.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Samukawa E, Fujihara S, Oura K, Iwama H, Yamana Y, Tadokoro T, Chiyo T, Kobayashi K, Morishita A, Nakahara M, Kobara H, Mori H, Okano K, Suzuki Y, Himoto T, Masaki T. Angiotensin receptor blocker telmisartan inhibits cell proliferation and tumor growth of cholangiocarcinoma through cell cycle arrest. Int J Oncol. 2017;51:1674–1684. doi: 10.3892/ijo.2017.4177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Salmani H, Hosseini M, Baghcheghi Y, Moradi-Marjaneh R, Mokhtari-Zaer A. Losartan modulates brain inflammation and improves mood disorders and memory impairment induced by innate immune activation: the role of PPAR-γ activation. Cytokine. 2020;125:154860. doi: 10.1016/j.cyto.2019.154860. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES