Next Article in Journal
The Early Prediction of Patient Outcomes in Acute Heart Failure: A Retrospective Study
Previous Article in Journal
Predictors of Five-Year Outcomes in Patients with Acute Coronary Syndromes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Navigating the Complexities of Cancer Treatment-Induced Hypertension

by
Jose Arriola-Montenegro
1,
John Roth
2 and
Maria L. Gonzalez Suarez
1,*
1
Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
2
Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
J. Cardiovasc. Dev. Dis. 2025, 12(6), 235; https://doi.org/10.3390/jcdd12060235
Submission received: 23 April 2025 / Revised: 11 June 2025 / Accepted: 13 June 2025 / Published: 19 June 2025

Abstract

Cancer therapy-induced hypertension (HTN) is an increasingly recognized complication associated with a wide range of anticancer agents, including vascular endothelial growth factor (VEGF) inhibitors, proteasome inhibitors, tyrosine kinase inhibitors, and alkylating agents. The pathogenesis of HTN in this setting is multifactorial, involving mechanisms such as endothelial dysfunction, nitric oxide (NO) suppression, sympathetic nervous system activation, and vascular remodeling. Additional factors, including paraneoplastic syndromes, poorly controlled pain, mood disturbances, and overlapping cardiovascular risk factors like obesity and diabetes, further contribute to the complexity of diagnosis and management. Despite its prevalence and clinical implications, cancer therapy-induced HTN is often addressed using general population guidelines, with limited oncology-specific protocols available. Accurate blood pressure measurement and individualized treatment plans are critical to optimize outcomes and avoid interruptions to cancer therapy. Antihypertensive agents such as angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARB), and calcium channel blockers have shown efficacy in both blood pressure control and, in some cases, oncologic outcomes. A multidisciplinary approach involving oncologists, cardiologists, and primary care providers is essential to navigate the interplay between cancer treatment and cardiovascular health. Ongoing research is needed to develop targeted guidelines and improve the long-term care of cancer patients affected by treatment-induced HTN.

1. Introduction

In recent decades, the number and diversity of cancer treatments have significantly increased, largely due to the development of targeted pharmacologic interventions [1]. While this expansion has greatly improved the prognosis for many cancer types, it has also revealed previously underrecognized toxicities, particularly those affecting the cardiovascular system [2]. Cancer therapy-induced hypertension (HTN) has emerged as a prominent and increasingly recognized cardiovascular complication. This includes various antineoplastic drug classes such as vascular endothelial growth factor (VEGF) inhibitors, proteasome inhibitors, and alkylating inhibitors, among others [2,3,4]. The International Cardio-Oncology Society (IC-OS), American Heart Association (AHA), and other professional organizations have recognized the urgent need to address cancer therapy-induced HTN [5,6]. As the use of these advanced treatments grows, understanding the relationship between HTN and cancer becomes more critical.
One complicating factor in cancer therapy-induced HTN is the frequent co-occurrence of acute kidney injury (AKI). The kidneys play a central role in blood pressure regulation through mechanisms like the renin–angiotensin–aldosterone system (RAAS), pressure natriuresis, and their interaction with the sympathetic nervous system [7,8,9,10]. Cancer patients are particularly vulnerable to renal damage, whether due to tumor invasion, treatment-related nephrotoxicity, hematopoietic stem cell transplant (HSCT) engraftment syndromes, tumor lysis syndrome (TLS), viral nephropathies, or common malignancy-related issues like sepsis and volume depletion [11,12,13,14]. Recurrent renal damage in this context can disrupt blood pressure (BP) regulation, further exacerbating HTN and adding complexity to the clinical picture [2].
In addition to the direct effects of cancer therapies, other malignancy-related factors further contribute to the development of HTN. These include poorly controlled pain, mood disorders such as anxiety and depression, and sleep disturbances, which are common amongst cancer patients [15,16,17,18]. Additional shared risk factors such as smoking, obesity, and diabetes further compound the risk [3,5,19]. Paraneoplastic HTN also represents another contributor to secondary HTN [20,21,22,23]. These diverse and interacting factors further complicate diagnosis and management of this condition.
Despite growing recognition, there remains a relative lack of dedicated studies and evidence-based guidelines to address this clinical entity. In practice, HTN in cancer patients is often treated empirically using standard guidelines for the general population [6]. However, the unique context of cancer and its treatments requires a more detailed approach. This review aims to highlight the pathophysiology of cancer therapy-induced HTN across various treatment classes, as well as current guidelines regarding its diagnostic approach and management of this unique clinical condition.

2. Paraneoplastic Hypertension

Paraneoplastic syndrome refers to cancer-associated signs caused by functional peptides, hormones, or immune cross-reactivity between tumor and normal host tissues [24]. Hypertension is a known paraneoplastic manifestation in various cancers, including renal tumors, neuroendocrine tumors, liver tumors, and, more rarely, ovarian tumors and pelvic teratomas [21,25,26,27,28].
Clear renal cell carcinoma is one of the most common tumors associated with paraneoplastic hypertension, affecting up to 64% of patients [21]. HTN in these cases is driven by the upregulation of the RAAS system, secretion of vasoactive peptides such as endothelin (ET-1), and deficiency of adrenomedullin, which can mediate BP [21,29,30]. Adrenomedullin is a potent vasodilator and natriuretic peptide, and studies have shown that its infusion leads to a reduction in BP [31,32]. Therefore, reduced levels of adrenomedullin may be consistent with the development of hypertension in these patients [33,34].
Pheochromocytomas and neuroendocrine tumors arise from chromaffin cells in the adrenal medulla, leading to unregulated catecholamine secretion. The classic triad of symptoms includes episodic headaches, sweating, and tachycardia, with patients developing sustained or paroxysmal HTN [35]. Hypertension occurs in about 80% of patients with pheochromocytoma, with half developing sustained HTN, 45% presenting with paroxysmal HTN, and 5–15% being normotensive [36,37,38,39]. Hypertension is primarily driven by norepinephrine release, and sustained HTN correlates with this release [40], while paroxysmal HTN is more common in patients with high plasma epinephrine levels [39].
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer [41,42], is also associated with HTN [26,43]. Studies show an increase in systolic blood pressure in HCC patients with concurrent diabetes mellitus [44]. Hypertension is a significant paraneoplastic phenomenon in HCC patients and is considered a potential predictor of survival [43]. The mechanism of HTN in HCC is thought to be caused by abnormalities in the RAAS system [45]. Previous reports have shown significant dysregulation of RAAS, including elevated renin levels, overproduction of angiotensin II, and a subsequent eight-to-tenfold increase in angiotensin II [26,45].

3. Etiologies of Cancer Treatment-Induced Hypertension

3.1. VEGF Signaling Pathway Inhibitors

VEGF signaling pathway inhibitors (VSPIs) are a widely utilized class of targeted pharmacotherapy in cancer treatment. These medications are clinically indicated for a variety of solid tumors, including colorectal carcinoma, renal cell carcinoma, non-small cell lung cancer, glioblastoma, and female reproductive cancers such as ovarian and cervical cancers [46,47,48,49,50,51,52]. The general, underlying principle is inhibition of angiogenesis primarily via disruption of the VEGF signaling cascade, a key regulator in the formation of new blood vessels [53]. The exact mechanism varies by agent, such as monoclonal antibodies targeting VEGF ligands in the case of bevacizumab as well as those against intracellular VEGF receptor tyrosine kinases, including sunitinib and sorafenib [54]. Collectively, these agents curtail tumor growth, invasion, and metastasis by reducing oxygen and nutrient delivery [55].
The efficacy of VSPIs has been repeatedly demonstrated in clinical trials, as in the case of AViSTAST, which investigated the use of bevacizumab in combination with traditional chemotherapy regimens in metastatic colorectal carcinoma [46]. Patients undergoing VSPI therapy displayed a significant increase in progression-free survival (PFS) and overall survival (OS) compared to standard chemotherapy regimens alone [46]. Likewise, increased PFS was noted with the use of sunitinib and sorafenib in metastatic renal cell carcinoma as compared to interferon-alpha [56,57]. In patients with non-small cell lung cancer, the addition of bevacizumab to carboplatin and paclitaxel resulted in significant improvements in OS as part of the ECOG 4599 trial [49]. Collectively, the results of these studies and similar investigations have repeatedly demonstrated the efficacy of VSPIs in the treatment of malignancy; however, they have also yielded significant insight into adverse events associated with these medications. HTN and proteinuria have notably emerged as frequent and clinically relevant complications associated with VSPI therapy, necessitating careful monitoring and management strategies [58,59].
HTN is a well-known and relatively common side effect in cancer patients treated with VSPIs, with an incidence of approximately 23.0%, although this can vary significantly based on the individual agent and dose [60,61,62]. Predisposing risk factors include patient factors such as female sex, advanced age (>60 years old), elevated BMI (>25 kg/m2), pre-existing HTN, and baseline renal dysfunction [63]. More recently, studies have correlated the development of HTN with low circulating plasma levels of VEGF-A and angiopoietin-2, as well as genetic variants of KCNAB1, which is involved in the production of voltage-gated potassium channels [64].
The pathophysiology of VSPI-induced HTN is complex and largely attributed to reduced nitric oxide (NO) production and vascular endothelial dysfunction [65]. VEGF-induced NO production promotes vasodilation, and disruption of this signaling cascade raises systemic vascular resistance, contributing to elevations in blood pressure [65]. Beyond systemic vasoconstriction, VSPIs are believed to influence renal sodium handling and glomerular filtration, further contributing to the development of HTN [65]. Consequently, patients who develop VSPI-induced HTN are at increased risk for a variety of complications. This includes stroke, myocardial infarction, and renal injury, all of which carry downstream effects on cancer treatment through dose reductions or even drug discontinuation [66].
Proteinuria is another well-characterized toxicity associated with VSPI therapy. Incidence has been estimated at 18.7%, although this can vary depending on the specific agent [67]. Patient-related risk factors include Asian ethnicity, diabetes, elevated baseline blood pressure, and prior nephrectomy, while medication-related factors include specific VSPI agents, increased dose, and treatment duration [68,69]. The underlying mechanism for VSPI-induced proteinuria involves damage to the glomerular endothelium and podocyte dysfunction. VEGF signaling is a key regulator for glomerular endothelial integrity, and disruption contributes to foot process effacement, which ultimately increases glomerular permeability [59,70]. In severe cases, VSPI-induced proteinuria may progress to nephrotic syndrome [71].

3.2. Proteasome Inhibitors

Proteasome inhibitors, such as bortezomib, carfilozib, and ixazomib, are commonly used in the treatment of multiple myeloma, primarily targeting the ubiquitin–proteasome system [72,73]. These anticancer drugs have been linked to high blood pressure, which affects approximately 10% of patients, although this varies by agent [74]. This toxicity is driven by multiple mechanisms, including angiotensin-induced HTN, aortic vascular remodeling, dysregulated NO homeostasis, and vasoconstriction [75,76,77]. Additionally, the accumulation of ubiquitinated proteins from proteasome inhibition leads to cellular apoptosis and endothelial damage, which can exacerbate cardiovascular adverse effects [73]. Moreover, numerous studies have demonstrated a strong association between carfilzomib and both new onset and worsening HTN [78,79,80], as well as other cardiac events such as heart failure, arrhythmias, and cardiac arrest [81].

3.3. Bruton’s Tyrosine Kinase (BTK) Inhibitors

BTK inhibitors are commonly used in the treatment of chronic lymphocytic leukemia, mantle cell lymphoma, and Waldenstrom macroglobulinemia [82,83,84]. However, these medications are well recognized for their association with the development of HTN [85,86,87]. This complication is attributed to multiple mechanisms, including VEGF inhibition, vascular fibrosis, cellular remodeling due to PI3K pathway inhibition, downregulation of NO, and endothelial dysfunction evidenced by an increase in plasma ET-1 and a decrease in plasma renin [88,89,90]. Studies have shown that the incidence of all-grade HTN is 23.4%, with 5.7% of patients experiencing high-grade HTN [91].

3.4. Rapidly Accelerated Fibrosarcoma B-Type (BRAF) and Mitogen-Activated Extracellular Signal-Regulated Kinase (MEK) Inhibitors

BRAF/MEK inhibitors target the Ras-RAF-MEK-ERK pathway and are commonly used for treatment of metastatic melanoma [92]. An important side effect of this dual treatment is the development of HTN, which is caused by the upregulation of CD47. This upregulation inhibits the NO/cyclic guanylyl monophosphate pathway, reducing NO production, which promotes vasoconstriction [92,93,94,95]. The incidence of HTN has been reported to be approximately 20% with BRAF/MEK inhibitors and 14% with BRAF inhibitor monotherapy [96]. Additional side effects due to Ras-RAF-MEK-ERK pathway inhibition include left ventricular systolic dysfunction, atrial arrhythmia, QT interval prolongation, and venous thromboembolism [96,97,98].

3.5. Radiation Therapy

Radiation therapy is an antineoplastic treatment that can predispose individuals to HTN through three main mechanisms: disruption of the carotid baroreceptor function, radiation nephropathy, and renal radiation-induced artery stenosis [99]. Specifically, neck radiation can lead to injury of the carotid arteries, causing atherosclerosis and fibrosis, which may reduce distensibility of the carotid sinus [100]. Impaired stretch-induced afferent carotid sinus nerve activity promotes chronic attenuation of both vagal and sympathetic baroreflex sensitivity. This dysfunction predisposes individuals to unrestrained sympathetic activation, which is evidenced as labile blood pressure, HTN, orthostatic intolerance, and tachycardia [101,102].
Radiation nephropathy is categorized in two stages based on the time after radiotherapy [103]. Acute radiation therapy occurs 6–18 months post-treatment. This is characterized by glomerular damage-related complications, such as proteinuria, edema, and azotemia [103,104]. Kidney biopsy findings in the acute phase primarily demonstrate vascular and glomerular changes: occluded capillary loops, loss of endothelial cells with subendothelial expansion, and mesangiolysis [105,106]. In contrast, chronic radiation nephropathy typically develops after 18 months and is associated with signs of chronic kidney disease, such as HTN, albuminuria, small atrophic kidneys, and anemia [103,107]. Common kidney biopsy features in the chronic stage include renal interstitial fibrosis, loss of nephron mass, and sclerosis of interlobular and arcuate arteries [106].
Renal radiation-induced artery stenosis is uncommon and typically affects the proximal segment of the renal artery. It typically develops after prolonged infradiaphragmatic radiation therapy and often presents as new-onset resistant HTN and elevated serum creatinine [108,109].

3.6. Alkylating Agents

Alkylating agents, such as cyclophosphamide, ifosfamide, busulfan, and cisplatin, are used in the treatment of hematologic malignancies (lymphoma and leukemia) and solid organ malignancies (testicular, brain, and ovarian cancers) [110,111]. These antineoplastic agents are associated with HTN via different mechanisms, including oxidative stress, cellular toxicity, endothelial damage, reduced VEGF concentrations, decreased NO bioavailability, direct vascular toxicity, and nephrotoxicity [75,112,113,114,115]. Cisplatin-based chemotherapy has been associated with an increased incidence of HTN as a late side effect for testicular cancer survivors [116]. Additionally, elevated blood pressure was common in cancer survivors after receiving treatment with ifosfamide [117]. Hypertension was evidenced in 25% to 36% of adults who received busulfan and in 58% of pediatric patients [118].

3.7. Platinum-Based Compounds

Platinum-based chemotherapies exert their neoplastic effects by inducing DNA cross-linking, thus inhibiting replication and promoting apoptosis [119]. These drugs are associated with the development of cancer treatment-induced HTN. This is thought to be multifactorial and primarily mediated through their direct effects upon the vasculature [119]. Vascular endothelial cells are vulnerable to the apoptotic effects of platinum derivatives, promoting cell death and a generalized inflammatory state [114]. Furthermore, platinum-containing drugs also promote endothelial dysfunction through a variety of mechanisms, including decreased production of NO [120]. There is growing evidence additionally linking the chronic neurotoxic effects of platinum agents to sympathetic dysfunction, further contributing to elevated blood pressure through downstream central nervous system-mediated reflexes [121]. Collectively, these effects promote increased systemic vascular resistance through widespread vasoconstriction, which induces the development of HTN [114,119,120,121,122]. Importantly, the hypertensive effects of platinum-based therapies may become chronic, as investigations have shown evidence of persistently elevated blood pressures even decades after exposure [122].

3.8. BCR-ABL Tyrosine Kinase Inhibitors

BCR-ABL tyrosine kinase inhibitors (TKIs) are selective, monoclonal antibodies that prevent cell division and ultimately promote apoptosis through downstream effects [123]. They are primarily utilized in the treatment of chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. These agents have been linked to various cardiovascular toxicities, which may be attributable to off-target inhibition of non-BCR-ABL TKIs [123]. However, the exact mechanisms behind these effects are still being investigated and are not always applicable to the entire class of medications. Up to 20–30% of cancer patients treated with ponatinib develop HTN thought to be attributed to inhibition of the VEGF system [124,125]. Nilotinib is another agent with an expansive cardiovascular risk profile, including the development of HTN. Studies have shown nilotinib to promote atherosclerosis as well as suppress endothelial cell proliferation, both of which likely contribute to the development of high blood pressure [126]. Given the distinct adverse event profiles of different TKIs, more investigation is required to better elucidate how other agents may contribute to the development of HTN [124].

3.9. Mammalian Target of Rapamycin Inhibitors

Mammalian target of rapamycin (mTOR) inhibitors, such as everolimus and sirolimus, are used as third-line treatment for renal cell carcinoma [56,127] and ovarian cancer [128]. In vitro studies have shown that mTOR inhibition decreases VEGF secretion, which may predispose patients to HTN [129,130]. Additionally, research suggests that mTOR inhibitors may induce HTN through increased oxidative stress and sympathetic activation, leading to afferent arteriolar vasoconstriction [131]. Everolimus has been associated with the development of all-grade HTN in patients with metastatic renal cell carcinoma [130]. Furthermore, the combination of everolimus and lenvatinib, a VEGF inhibitor, was associated with all-grade HTN in 41% of patients and high-grade HTN in 14% of patients [130]. Moreover, mTOR inhibitors impair glucose and lipid metabolism and contribute to abdominal obesity, which may further increase the cardiovascular risk associated with these agents [132,133].

3.10. Anti-Androgen Therapy

Anti-androgen therapy is a cornerstone of prostate cancer treatment and includes multiple classes of medications, such as androgen receptor antagonists such as cyproterone acetate, flutamide, enzalutamide, and bicalutamide, as well as 17α-hydroxylase/C17,20-lyase (CYP17) inhibitors that block androgen synthesis, such as abiraterone [134]. These agents have well-established metabolic and cardiovascular risk profiles, including the development of HTN. Abiraterone inhibits production of both androgens and cortisol through its effect upon CYP17 enzymes [134]. With the loss of the cortisol-mediated negative feedback loop, subsequent overexpression of adrenocorticotropic hormone results in a buildup of mineralocorticoid precursors and development of secondary HTN [134]. The exact mechanism by which androgen receptor antagonists are associated with elevated blood pressure is less clear. A higher incidence of HTN was noted in those prostate cancer patients treated with enzalutamide [135]. Similarly, in the context of gender-affirming hormonal therapy, cyproterone acetate has been associated with increased blood pressure and HTN onset [136]. More investigation is needed to fully elucidate the cardiovascular effects of these agents.

3.11. Adjuvant Therapies

Many cancer patients receive adjuvant therapies alongside chemotherapy as part of their treatment plan. These therapies may contribute to the development of HTN or exacerbate pre-existing HTN [118,137,138]. Medications such as erythropoiesis-stimulating agents (ESAs), glucocorticoids, nonsteroidal anti-inflammatory drugs (NSAIDs), and calcineurin inhibitors are commonly involved and are known to cause HTN [137,138]. Therefore, it is essential to carefully monitor patients when these drugs are included in the cancer treatment regimen, especially when used in combination with antineoplastic agents known to increase blood pressure.
ESAs have been associated with an increased risk for HTN in multiple clinical trials [137]. Various mechanisms have been identified, including increased blood viscosity, arterial remodeling, and increased vascular resistance to the vasodilator effects of NO, which predisposes patients to vasoconstriction [139,140].
Glucocorticoids are known for their side effects, including water and sodium retention, which can lead to a rise in blood pressure [141]. Glucocorticoid-induced HTN has been reported in up to 13% of patients [142].
NSAIDs are recognized for their significant association with HTN [143]. These side effects are primarily driven by their inhibitory effect on the production of vasodilatory prostaglandins and retention of water and salt [144]. Studies have shown an increase in supine mean blood pressure by 5.0 mmHg [143].
Through the inhibition of T-cell function, calcineurin inhibitors prevent the rejection of transplanted solid organs and graft-versus-host disease in the context of allogeneic bone marrow transplantation. These medications have been shown to be associated with HTN. Mechanisms include activation of RAAS and the sympathetic nervous system, increased proximal tubule sodium reabsorption, endothelial dysfunction via stimulation of endothelin-1, inhibition of NO production, and increased oxidative stress [145,146]. Additionally, these medications may predispose patients to nephrotoxicity, which can contribute to prohypertensive effects [145]. Some 30–80% of patients treated with calcineurin inhibitors develop HTN [147,148].
A graphical summary of the key mechanisms involved in cancer treatment-induced hypertension is provided in Figure 1. Additionally, Table 1 outlines the mechanisms, representative medications, and relative incidence of all-grade hypertension across major oncologic drug classes, while Table 2 highlights the hypertensive impact of radiation and adjuvant therapies. Together, these tools aim to support risk stratification and clinical decision-making in this complex population.

4. Diagnosis of Hypertension in Cancer Patients

Accurate measurement of BP is crucial for diagnosing and managing HTN, particularly in cancer patients. These patients may experience pain or anxiety during clinic visits or be taking medications such as NSAIDs, ESAs, and corticosteroids, which can interfere with BP readings [6,118].
For accurate measurement, patients should be seated quietly and at rest for 3 to 5 min before their BP is taken. The measurement should be carried out in a quiet room with the patient’s legs resting on the floor (uncrossed) and their back properly supported. The arm used for BP measurement should be at heart level, with a correctly sized, calibrated cuff placed on a bare arm. Additionally, the patient should have an empty bladder and avoid caffeine or smoking for at least 30 min before the measurement [6,118,158,159].
During the clinic visit, at least three BP readings should be obtained and averaged. If any readings are elevated, they should be verified on at least one additional measurement before diagnosing HTN [158]. Automated office BP measurement can be a useful tool for obtaining multiple readings in a single visit and can be performed with or without the presence of a provider [160]. For initial BP readings ≥ 120/70 mmHg, 24-hour ambulatory BP monitoring is recommended to confirm the diagnosis [160]. However, this method is not always feasible to perform more than twice a year for elevated readings [161]. As an alternative, home BP monitoring requires patients to measure BP twice a day for at least five consecutive days. This method is often preferred by both patients and clinicians, as it allows for close monitoring of BP, facilitating medication adjustments and helping identify white coat hypertension [162].
The 2017 American College of Cardiology/American Heart Association (ACC/AHA) Clinical Practice Guidelines and the 2018 ESC/European Society of Hypertension Guidelines are commonly used to diagnose and grade HTN in cancer patients. Hypertension is diagnosed when office-based or average home BP measurements are ≥130/80 mmHg or when the average BP on ambulatory blood pressure monitoring (ABPM) is ≥125/75 mmHg [158,163,164].
It is essential to measure baseline BP before starting prohypertensive anticancer therapies, as some patients may experience a significant increase in BP during treatment, requiring prompt initiation or adjustment of antihypertensive therapy. The National Cancer Institute Investigational Drug Steering Committee recommends targeting a BP of <140/90 mmHg before starting VSPI therapy [165].

5. Blood Pressure Goal in Patients with Cancer

Patients with an estimated 10-year atherosclerotic cardiovascular disease (ASCVD) risk of ≥10% or those with additional cardiovascular comorbidities such as type 2 diabetes mellitus, chronic kidney disease, stroke, or peripheral vascular disease should aim for a BP goal of <130/80 mmHg. Patients with an estimated 10-year ASCVD risk of ≤10% and without additional cardiovascular comorbidities should aim for a BP goal of <140/90 mmHg [158].
For asymptomatic patients with metastatic cancer and an expected survival of 1–3 years, the target BP may be increased to 140–159/90–99 mmHg. However, if BP rises above >160/100 mmHg, treatment should be initiated in all patients to prevent life-threatening complications [5,99]. Additionally, prohypertensive anticancer agents should be withheld if BP rises above 180/110 mmHg and should not be restarted until BP is controlled to <160/110 mmHg [4,5]. If a hypertensive emergency occurs, consideration of permanent discontinuation of prohypertensive anticancer agents should be made [99].

6. Management of Cancer Therapy-Related HTN

6.1. Lifestyle Modifications

Patients should be counseled on lifestyle changes that can help improve BP control. This includes limiting sodium intake, maintaining a healthy weight, reducing caffeine and alcohol consumption, avoiding NSAIDs, and increasing physical activity [6,166,167]. Providers should also address the impact of concomitant therapies that may elevate BP during anticancer therapy, such as corticosteroids or ESAs [168]. Additionally, managing pain, minimizing anxiety, and evaluating and treating comorbidities (e.g., obstructive sleep apnea or diabetes mellitus) that may contribute to high BP are essential steps in effective HTN management [6,44,99].

6.2. Antihypertensive Medications

Currently, there are no specific antihypertensive guidelines for cancer patients undergoing anticancer therapies, and antihypertensive management should follow the general population’s guidelines [169]. However, further recommendations emphasize that HTN management should be a multidisciplinary approach, including cardiologists, oncologists, nurses, primary care physicians, patients, and their families [6].
The choice of antihypertensive agent should be individualized based on the type of anticancer treatment. For instance, non-dihydropyridine calcium channel blockers (e.g., diltiazem and verapamil) should be avoided in patients receiving therapies metabolized by P-glycoprotein and cytochrome P450 3A4 [170] as well as sunitinib and sorafenib due to known interactions [171]. First-line treatments typically include angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), dihydropyridine calcium channel blockers (CCB), or diuretics [172,173,174,175,176].
ACE inhibitors and ARBs are considered the first-line treatment for anti-VEGF-induced HTN [177]. This is due to their renoprotective effects, which are particularly beneficial for diabetic patients at risk of proteinuria during VEGF inhibition therapy [178]. These agents are particularly effective in managing mild increases in BP (10–15 mmHg) during initiation of anticancer treatment [178]. Experimental studies have shown that losartan and captopril reduce VEGF expression in renal tumors, suppress tumor size, and shrink lung metastases in renal cell carcinoma patients [179]. Furthermore, ACE inhibitors and ARBs have been associated with improved survival in metastatic renal cell carcinoma patients [180] and lower colorectal cancer risk following a negative baseline colonoscopy [181].
Dihydropyridine CCBs such as amlodipine have been associated with improvements in de novo HTN and worsening HTN in patients undergoing treatment with bevacizumab for advanced or metastatic non-small cell lung carcinoma, colorectal cancer, or ovarian cancer [182]. Additionally, CCBs and potassium-sparing agents were associated with the largest reductions in BP in patients on anti-VEGF tyrosine kinase inhibitor therapy for metastatic renal cell cancer [174]. Thiazide diuretics are an acceptable alternative. However, caution must be considered in patients at risk of volume depletion secondary to chemotherapy [6].
Sodium–glucose cotransporter 2 (SGLT2) inhibitors are emerging as a potential adjunctive therapy for cancer therapy-induced HTN. Beyond their glycemic benefits, SGLT2 inhibitors have demonstrated modest blood pressure-lowering effects and significant cardiorenal protection [183]. SGLT2 inhibitors have been shown to reduce endothelial dysfunction through a variety of mechanisms, which may be particularly advantageous in those treated with VSPIs or other classes known to disrupt endothelial homeostasis [184]. Moreover, empagliflozin has been shown to reduce sunitinib-induced HTN in addition to other treatment-related cardiac toxicities [185]. As cancer patients have often been excluded from randomized controlled trials involving SGLT2 inhibitors, more research is needed to explore the cardiovascular benefits of these medications in cancer patients.
To enhance clinical applicability, we have incorporated a practical flowchart (Figure 2) that outlines a stepwise approach to managing hypertension in patients receiving VSPIs, integrating both pharmacologic and non-pharmacologic interventions. This aims to guide clinicians in tailoring management strategies based on oncologic regimen and patient-specific risk factors. Additionally, we acknowledge critical gaps in the field, including the absence of standardized blood pressure monitoring protocols during cancer therapy, limited comparative studies evaluating antihypertensive strategies in this population, and the lack of cardio-oncology-specific BP targets in current guidelines. These gaps underscore the urgent need for further research to optimize cardiovascular care in oncology patients.

7. Conclusions

Cancer therapy-induced hypertension is a common and clinically significant complication that requires early recognition and individualized management. While current treatment often follows general hypertension guidelines, the unique risks associated with cancer therapies necessitate a tailored, multidisciplinary approach. Further research is essential to develop dedicated guidelines and improve outcomes for cancer patients affected by treatment-related hypertension.

Author Contributions

Conceptualization, J.A.-M. and M.L.G.S.; methodology, J.A.-M., J.R. and M.L.G.S.; validation, J.A.-M., J.R. and M.L.G.S.; investigation, J.A.-M., J.R. and M.L.G.S.; resources, J.A.-M. and M.L.G.S.; writing—original draft preparation, J.A.-M., J.R. and M.L.G.S.; writing—review and editing, J.A.-M. and M.L.G.S.; visualization, J.A.-M.; supervision, M.L.G.S.; project administration, J.A.-M. and M.L.G.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data was generated for this review.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Scott, E.C.; Baines, A.C.; Gong, Y.; Moore, R.; Pamuk, G.E.; Saber, H.; Subedee, A.; Thompson, M.D.; Xiao, W.; Pazdur, R.; et al. Trends in the approval of cancer therapies by the FDA in the twenty-first century. Nat. Rev. Drug Discov. 2023, 22, 625–640. [Google Scholar] [CrossRef]
  2. Van Leeuwen, M.T.; Luu, S.; Gurney, H.; Brown, M.R.; Pearson, S.A.; Webber, K.; Hunt, L.; Soojung, H.; Delaney, G.P.; Vajdic, C.M. Cardiovascular Toxicity of Targeted Therapies for Cancer: An Overview of Systematic Reviews. JNCI Cancer Spectr. 2020, 4, pkaa076. [Google Scholar] [CrossRef] [PubMed]
  3. Bloom, M.W.; Hamo, C.E.; Cardinale, D.; Ky, B.; Nohria, A.; Baer, L.; Skopicki, H.; Lenihan, D.J.; Gheorghiade, M.; Lyon, A.R.; et al. Cancer Therapy-Related Cardiac Dysfunction and Heart Failure: Part 1: Definitions, Pathophysiology, Risk Factors, and Imaging. Circ. Heart Fail. 2016, 9, e002661. [Google Scholar] [CrossRef]
  4. Herrmann, J.; Lenihan, D.; Armenian, S.; Barac, A.; Blaes, A.; Cardinale, D.; Carver, J.; Dent, S.; Ky, B.; Lyon, A.R.; et al. Defining cardiovascular toxicities of cancer therapies: An International Cardio-Oncology Society (IC-OS) consensus statement. Eur. Heart J. 2022, 43, 280–299. [Google Scholar] [CrossRef] [PubMed]
  5. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
  6. Cohen, J.B.; Brown, N.J.; Brown, S.A.; Dent, S.; van Dorst, D.C.H.; Herrmann, S.M.; Lang, N.N.; Oudit, G.Y.; Touyz, R.M.; American Heart Association Council on Hypertension; et al. Cancer Therapy-Related Hypertension: A Scientific Statement From the American Heart Association. Hypertension 2023, 80, e46–e57. [Google Scholar] [CrossRef] [PubMed]
  7. Sparks, M.A.; Crowley, S.D.; Gurley, S.B.; Mirotsou, M.; Coffman, T.M. Classical Renin-Angiotensin system in kidney physiology. Compr. Physiol. 2014, 4, 1201–1228. [Google Scholar] [CrossRef]
  8. Carlström, M.; Wilcox, C.S.; Arendshorst, W.J. Renal autoregulation in health and disease. Physiol. Rev. 2015, 95, 405–511. [Google Scholar] [CrossRef]
  9. Ivy, J.R.; Bailey, M.A. Pressure natriuresis and the renal control of arterial blood pressure. J. Physiol. 2014, 592, 3955–3967. [Google Scholar] [CrossRef]
  10. Wadei, H.M.; Textor, S.C. The role of the kidney in regulating arterial blood pressure. Nat. Rev. Nephrol. 2012, 8, 602–609. [Google Scholar] [CrossRef]
  11. Rosner, M.H.; Perazella, M.A. Acute Kidney Injury in Patients with Cancer. N. Engl. J. Med. 2017, 376, 1770–1781. [Google Scholar] [CrossRef] [PubMed]
  12. Malyszko, J.; Kozlowska, K.; Kozlowski, L.; Malyszko, J. Nephrotoxicity of anticancer treatment. Nephrol. Dial. Transplant. 2017, 32, 924–936. [Google Scholar] [CrossRef] [PubMed]
  13. Troxell, M.L.; Higgins, J.P.; Kambham, N. Antineoplastic Treatment and Renal Injury: An Update on Renal Pathology Due to Cytotoxic and Targeted Therapies. Adv. Anat. Pathol. 2016, 23, 310–329. [Google Scholar] [CrossRef]
  14. Bridoux, F.; Cockwell, P.; Glezerman, I.; Gutgarts, V.; Hogan, J.J.; Jhaveri, K.D.; Joly, F.; Nasr, S.H.; Sawinski, D.; Leung, N. Kidney injury and disease in patients with haematological malignancies. Nat. Rev. Nephrol. 2021, 17, 386–401. [Google Scholar] [CrossRef] [PubMed]
  15. McMillan, S.C.; Tofthagen, C.; Morgan, M.A. Relationships among pain, sleep disturbances, and depressive symptoms in outpatients from a comprehensive cancer center. Oncol. Nurs. Forum 2008, 35, 603–611. [Google Scholar] [CrossRef]
  16. Bortolato, B.; Hyphantis, T.N.; Valpione, S.; Perini, G.; Maes, M.; Morris, G.; Kubera, M.; Köhler, C.A.; Fernandes, B.S.; Stubbs, B.; et al. Depression in cancer: The many biobehavioral pathways driving tumor progression. Cancer Treat. Rev. 2017, 52, 58–70. [Google Scholar] [CrossRef]
  17. Carey, R.M.; Calhoun, D.A.; Bakris, G.L.; Brook, R.D.; Daugherty, S.L.; Dennison-Himmelfarb, C.R.; Egan, B.M.; Flack, J.M.; Gidding, S.S.; Judd, E.; et al. Resistant Hypertension: Detection, Evaluation, and Management: A Scientific Statement From the American Heart Association. Hypertension 2018, 72, e53–e90. [Google Scholar] [CrossRef]
  18. Palagini, L.; Bruno, R.M.; Gemignani, A.; Baglioni, C.; Ghiadoni, L.; Riemann, D. Sleep loss and hypertension: A systematic review. Curr. Pharm. Des. 2013, 19, 2409–2419. [Google Scholar] [CrossRef]
  19. Johnson, C.B.; Davis, M.K.; Law, A.; Sulpher, J. Shared Risk Factors for Cardiovascular Disease and Cancer: Implications for Preventive Health and Clinical Care in Oncology Patients. Can. J. Cardiol. 2016, 32, 900–907. [Google Scholar] [CrossRef]
  20. Shah, M.H.; Goldner, W.S.; Benson, A.B.; Bergsland, E.; Blaszkowsky, L.S.; Brock, P.; Chan, J.; Das, S.; Dickson, P.V.; Fanta, P.; et al. Neuroendocrine and Adrenal Tumors, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc Netw. 2021, 19, 839–868. [Google Scholar] [CrossRef]
  21. Gupta, S.; Nichols, P.; Lohse, C.M.; Kosari, F.; Kattah, A.G.; Harris, F.R.; Karagouga, G.; Mehra, R.; Fine, S.M.; Reuter, V.E.; et al. Renin Production by Juxtaglomerular Cell Tumors and Clear Cell Renal Cell Carcinoma and the Role of Angiotensin Signaling Inhibitors. Mayo Clin. Proc. 2022, 97, 2050–2064. [Google Scholar] [CrossRef] [PubMed]
  22. Pappachan, J.M.; Tun, N.N.; Arunagirinathan, G.; Sodi, R.; Hanna, F.W.F. Pheochromocytomas and Hypertension. Curr. Hypertens. Rep. 2018, 20, 3. [Google Scholar] [CrossRef] [PubMed]
  23. Boudreaux, J.P.; Klimstra, D.S.; Hassan, M.M.; Woltering, E.A.; Jensen, R.T.; Goldsmith, S.J.; Nutting, C.; Bushnell, D.L.; Caplin, M.E.; Yao, J.C.; et al. The NANETS Consensus Guideline for the Diagnosis and Management of Neuroendocrine Tumors: Well-Differentiated Neuroendocrine Tumors of the Jejunum, Ileum, Appendix, and Cecum. Pancreas 2010, 39, 753–766. [Google Scholar] [CrossRef] [PubMed]
  24. Pelosof, L.C.; Gerber, D.E. Paraneoplastic syndromes: An approach to diagnosis and treatment. Mayo Clin. Proc. 2010, 85, 838–854. [Google Scholar] [CrossRef] [PubMed]
  25. Haq, N.; Taherian, M.; Rout, P. Adrenal Cortical Nodular Hyperplasia. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2025. [Google Scholar]
  26. Kew, M.C.; Leckie, B.J.; Greeff, M.C. Arterial hypertension as a paraneoplastic phenomenon in hepatocellular carcinoma. Arch. Intern. Med. 1989, 149, 2111–2113. [Google Scholar] [CrossRef]
  27. Pursell, R.N.; Quinlan, P.M. Secondary hypertension due to a renin-producing teratoma. Am. J. Hypertens. 2003, 16, 592–595. [Google Scholar] [CrossRef]
  28. Alrobaiq, B.M.; Alharbi, R.S.; Alhoshan, F.S.; Alnasyan, M.A.; Alahideb, A.; Omair, A. Hypertension and Ovarian Cancer: A Case-Control Study in Saudi Arabia. Cureus 2023, 15, e35294. [Google Scholar] [CrossRef]
  29. Sauzeau, V.; Le Mellionnec, E.; Bertoglio, J.; Scalbert, E.; Pacaud, P.; Loirand, G. Human urotensin II-induced contraction and arterial smooth muscle cell proliferation are mediated by RhoA and Rho-kinase. Circ. Res. 2001, 88, 1102–1104. [Google Scholar] [CrossRef]
  30. Pflug, B.R.; Zheng, H.; Udan, M.S.; D’Antonio, J.M.; Marshall, F.F.; Brooks, J.D.; Nelson, J.B. Endothelin-1 promotes cell survival in renal cell carcinoma through the ET(A) receptor. Cancer Lett. 2007, 246, 139–148. [Google Scholar] [CrossRef]
  31. Hu, W.; Zhou, P.H.; Zhang, X.B.; Xu, C.G.; Wang, W. Plasma concentrations of adrenomedullin and natriuretic peptides in patients with essential hypertension. Exp. Ther. Med. 2015, 9, 1901–1908. [Google Scholar] [CrossRef]
  32. Wang, H.Y.; Wang, F.Z.; Chang, R.; Wang, Q.; Liu, S.Y.; Cheng, Z.X.; Gao, Q.; Zhou, H.; Zhou, Y.-B. Adrenomedullin Improves Hypertension and Vascular Remodeling partly through the Receptor-Mediated AMPK Pathway in Rats with Obesity-Related Hypertension. Int. J. Mol. Sci. 2023, 24, 3943. [Google Scholar] [CrossRef] [PubMed]
  33. Iring, A.; Jin, Y.J.; Albarrán-Juárez, J.; Siragusa, M.; Wang, S.; Dancs, P.T.; Nakayama, A.; Tonack, S.; Chen, M.; Künne, C.; et al. Shear stress-induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J. Clin. Investig. 2019, 129, 2775–2791. [Google Scholar] [CrossRef]
  34. Mogi, M. Effect of adrenomedullin on obesity-related hypertension. Hypertens. Res. 2024, 47, 2221–2222. [Google Scholar] [CrossRef] [PubMed]
  35. Stein, P.P.; Black, H.R. A simplified diagnostic approach to pheochromocytoma. A review of the literature and report of one institution’s experience. Medicine 1991, 70, 46–66. [Google Scholar] [CrossRef]
  36. Manger, W.M. The protean manifestations of pheochromocytoma. Horm. Metab. Res. 2009, 41, 658–663. [Google Scholar] [CrossRef]
  37. Zelinka, T.; Eisenhofer, G.; Pacak, K. Pheochromocytoma as a catecholamine producing tumor: Implications for clinical practice. Stress 2007, 10, 195–203. [Google Scholar] [CrossRef]
  38. Zuber, S.M.; Kantorovich, V.; Pacak, K. Hypertension in pheochromocytoma: Characteristics and treatment. Endocrinol. Metab. Clin. N. Am. 2011, 40, 295–311+vii. [Google Scholar] [CrossRef] [PubMed]
  39. Manger, W.M.; Eisenhofer, G. Pheochromocytoma: Diagnosis and management update. Curr. Hypertens. Rep. 2004, 6, 477–484. [Google Scholar] [CrossRef]
  40. O’Rourke, M.F.; Staessen, J.A.; Vlachopoulos, C.; Duprez, D.; Plante, G.E. Clinical applications of arterial stiffness; definitions and reference values. Am. J. Hypertens. 2002, 15, 426–444. [Google Scholar] [CrossRef]
  41. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
  42. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef]
  43. Hu, Y.F.; Chen, T.C.; Chau, G.Y.; Yang, T.L.; Liu, C.J.; Chen, M.H.; Chang, P.M.-H.; Chen, T.-J.; Hsiao, M.; Huang, C.-Y.F.; et al. Baseline hypertension: New insight into the potential predictors of survival in patients with hepatocellular carcinoma. Int. J. Cardiol. 2013, 168, 2979–2981. [Google Scholar] [CrossRef]
  44. Zhang, W.S.; Li, X.O.; Zhang, H.; Gao, C.; Fang, L.; Yang, H.Y. Increased Level of Systolic Blood Pressure in Hepatocellular Carcinoma Patients with Diabetes Mellitus. Diabetes Metab. Syndr. Obes. 2020, 13, 1979–1988. [Google Scholar] [CrossRef]
  45. Yoshiji, H.; Noguchi, R.; Ikenaka, Y.; Kaji, K.; Aihara, Y.; Fukui, H. Impact of renin-angiotensin system in hepatocellular carcinoma. Curr. Cancer Drug Targets 2011, 11, 431–441. [Google Scholar] [CrossRef] [PubMed]
  46. Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E.; et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef]
  47. Chinot, O.L.; Wick, W.; Mason, W.; Henriksson, R.; Saran, F.; Nishikawa, R.; Carpentier, A.F.; Hoang-Xuan, K.; Kavan, P.; Cernea, D.; et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 2014, 370, 709–722. [Google Scholar] [CrossRef] [PubMed]
  48. Iacovelli, R.; Sternberg, C.N.; Porta, C.; Verzoni, E.; de Braud, F.; Escudier, B.; Procopio, G. Inhibition of the VEGF/VEGFR pathway improves survival in advanced kidney cancer: A systematic review and meta-analysis. Curr. Drug Targets 2015, 16, 164–170. [Google Scholar] [CrossRef] [PubMed]
  49. Sandler, A.; Gray, R.; Perry, M.C.; Brahmer, J.; Schiller, J.H.; Dowlati, A.; Lilenbaum, R.; Johnson, D.H. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N. Engl. J. Med. 2006, 355, 2542–2550. [Google Scholar] [CrossRef]
  50. Oura, K.; Morishita, A.; Tadokoro, T.; Fujita, K.; Tani, J.; Kobara, H. Immune Microenvironment and the Effect of Vascular Endothelial Growth Factor Inhibition in Hepatocellular Carcinoma. Int. J. Mol. Sci. 2024, 25, 13590. [Google Scholar] [CrossRef]
  51. Murphy, A.D.; Morgan, R.D.; Clamp, A.R.; Jayson, G.C. The role of vascular endothelial growth factor inhibitors in the treatment of epithelial ovarian cancer. Br. J. Cancer 2022, 126, 851–864. [Google Scholar] [CrossRef]
  52. Kamat, A.A.; Merritt, W.M.; Coffey, D.; Lin, Y.G.; Patel, P.R.; Broaddus, R.; Nurgent, E.; Han, L.Y.; Landen Jr, C.N.; Spannuth, W.A.; et al. Clinical and biological significance of vascular endothelial growth factor in endometrial cancer. Clin. Cancer Res. 2007, 13, 7487–7495. [Google Scholar] [CrossRef] [PubMed]
  53. Ferrara, N. Vascular Endothelial Growth Factor: Basic Science and Clinical Progress. Endocr. Rev. 2004, 25, 581–611. [Google Scholar] [CrossRef]
  54. Wilhelm, S.M.; Carter, C.; Tang, L.; Wilkie, D.; McNabola, A.; Rong, H.; Chen, C.; Zhang, X.; Vincent, P.; McHugh, M.; et al. BAY 43-9006 Exhibits Broad Spectrum Oral Antitumor Activity and Targets the RAF/MEK/ERK Pathway and Receptor Tyrosine Kinases Involved in Tumor Progression and Angiogenesis. Cancer Res. 2004, 64, 7099–7109. [Google Scholar] [CrossRef]
  55. Kerbel, R.S. Tumor angiogenesis. N. Engl. J. Med. 2008, 358, 2039–2049. [Google Scholar] [CrossRef] [PubMed]
  56. Escudier, B.; Eisen, T.; Stadler, W.M.; Szczylik, C.; Oudard, S.; Siebels, M.; Negrier, S.; Chevreau, C.; Solska, E.; Desai, A.A.; et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N. Engl. J. Med. 2007, 356, 125–134. [Google Scholar] [CrossRef] [PubMed]
  57. Motzer, R.J.; Hutson, T.E.; Tomczak, P.; Michaelson, M.D.; Bukowski, R.M.; Rixe, O.; Oudard, S.; Negrier, S.; Szcylik, C.; Kim, S.T.; et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N. Engl. J. Med. 2007, 356, 115–124. [Google Scholar] [CrossRef]
  58. Versmissen, J.; Mirabito Colafella, K.M.; Koolen, S.L.W.; Danser, A.H.J. Vascular Cardio-Oncology: Vascular Endothelial Growth Factor inhibitors and hypertension. Cardiovasc. Res. 2019, 115, 904–914. [Google Scholar] [CrossRef]
  59. Izzedine, H.; Massard, C.; Spano, J.P.; Goldwasser, F.; Khayat, D.; Soria, J.C. VEGF signalling inhibition-induced proteinuria: Mechanisms, significance, and management. Eur. J. Cancer 2010, 46, 439–448. [Google Scholar] [CrossRef]
  60. Liu, B.; Ding, F.; Liu, Y.; Xiong, G.; Lin, T.; He, D.; Zhang, Y.; Zhang, D.; Wei, G. Incidence and risk of hypertension associated with vascular endothelial growth factor receptor tyrosine kinase inhibitors in cancer patients: A comprehensive network meta-analysis of 72 randomized controlled trials involving 30,013 patients. Oncotarget 2016, 7, 67661–67673. [Google Scholar] [CrossRef]
  61. van Dorst, D.C.H.; Kabadayi, S.; Oomen-de Hoop, E.; Danser, A.H.J.; Mathijssen, R.H.J.; Versmissen, J. Treatment and Implications of Vascular Endothelial Growth Factor Inhibitor-Induced Blood Pressure Rise: A Clinical Cohort Study. J. Am. Heart Assoc. 2023, 12, e028050. [Google Scholar] [CrossRef]
  62. Moslehi, J.J. Cardiovascular Toxic Effects of Targeted Cancer Therapies. N. Engl. J. Med. 2016, 375, 1457–1467. [Google Scholar] [CrossRef] [PubMed]
  63. Hamnvik, O.P.; Choueiri, T.K.; Turchin, A.; McKay, R.R.; Goyal, L.; Davis, M.; Kaymakcalan, D.M.; Williams, J.S. Clinical risk factors for the development of hypertension in patients treated with inhibitors of the VEGF signaling pathway. Cancer 2015, 121, 311–319. [Google Scholar] [CrossRef] [PubMed]
  64. Quintanilha, J.C.F.; Kelly, W.K.; Innocenti, F. Contribution of plasma levels of VEGF-A and angiopoietin-2 in addition to a genetic variant in KCNAB1 to predict the risk of bevacizumab-induced hypertension. Pharmacogenomics J. 2024, 24, 22. [Google Scholar] [CrossRef] [PubMed]
  65. Pandey, A.K.; Singhi, E.K.; Arroyo, J.P.; Ikizler, T.A.; Gould, E.R.; Brown, J.; Beckman, J.A.; Harrison, D.G.; Moslehi, J. Mechanisms of VEGF (Vascular Endothelial Growth Factor) Inhibitor-Associated Hypertension and Vascular Disease. Hypertension 2018, 71, e1–e8. [Google Scholar] [CrossRef]
  66. Li, J.; Gu, J. Cardiovascular Toxicities with Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitors in Cancer Patients: A Meta-Analysis of 77 Randomized Controlled Trials. Clin. Drug Investig. 2018, 38, 1109–1123. [Google Scholar] [CrossRef]
  67. Zhang, Z.F.; Wang, T.; Liu, L.H.; Guo, H.Q. Risks of proteinuria associated with vascular endothelial growth factor receptor tyrosine kinase inhibitors in cancer patients: A systematic review and meta-analysis. PLoS ONE 2014, 9, e90135. [Google Scholar] [CrossRef]
  68. Kato, T.; Mizuno, R.; Miyake, H. Prevalence and management of proteinuria associated with vascular endothelial growth factor receptor-targeted tyrosine kinase inhibitor treatment in advanced renal cell carcinoma, hepatocellular carcinoma, and thyroid cancer. Int. J. Urol. 2024, 31, 465–474. [Google Scholar] [CrossRef]
  69. 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. [Google Scholar] [CrossRef]
  70. Eremina, V.; Quaggin, S.E. The role of VEGF-A in glomerular development and function. Curr. Opin. Nephrol. Hypertens. 2004, 13, 9–15. [Google Scholar] [CrossRef]
  71. Abbas, A.; Mirza, M.M.; Ganti, A.K.; Tendulkar, K. Renal Toxicities of Targeted Therapies. Target. Oncol. 2015, 10, 487–499. [Google Scholar] [CrossRef]
  72. Park, J.; Cho, J.; Song, E.J. Ubiquitin-proteasome system (UPS) as a target for anticancer treatment. Arch. Pharm. Res. 2020, 43, 1144–1161. [Google Scholar] [CrossRef] [PubMed]
  73. Hasinoff, B.B.; Patel, D.; Wu, X. Molecular Mechanisms of the Cardiotoxicity of the Proteasomal-Targeted Drugs Bortezomib and Carfilzomib. Cardiovasc. Toxicol. 2017, 17, 237–250. [Google Scholar] [CrossRef] [PubMed]
  74. Georgiopoulos, G.; Makris, N.; Laina, A.; Theodorakakou, F.; Briasoulis, A.; Trougakos, I.P.; Dismopoulos, M.-A.; Kastritis, E.; Stametelopoulos, K. Cardiovascular Toxicity of Proteasome Inhibitors: Underlying Mechanisms and Management Strategies: JACC: Cardio Oncology State-of-the-Art Review. JACC Cardio Oncol. 2023, 5, 1–21. [Google Scholar] [CrossRef]
  75. Mohammed, T.; Singh, M.; Tiu, J.G.; Kim, A.S. Etiology and management of hypertension in patients with cancer. Cardio-oncology 2021, 7, 14. [Google Scholar] [CrossRef]
  76. Chari, A.; Hajje, D. Case series discussion of cardiac and vascular events following carfilzomib treatment: Possible mechanism, screening, and monitoring. BMC Cancer 2014, 14, 915. [Google Scholar] [CrossRef]
  77. Wu, P.; Oren, O.; Gertz, M.A.; Yang, E.H. Proteasome Inhibitor-Related Cardiotoxicity: Mechanisms, Diagnosis, and Management. Curr. Oncol. Rep. 2020, 22, 66. [Google Scholar] [CrossRef] [PubMed]
  78. Dimopoulos, M.A.; Goldschmidt, H.; Niesvizky, R.; Joshua, D.; Chng, W.J.; Oriol, A.; Orlowski, R.Z.; Ludwig, H.; Facon, T.; Hajek, R.; et al. Carfilzomib or bortezomib in relapsed or refractory multiple myeloma (ENDEAVOR): An interim overall survival analysis of an open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 1327–1337. [Google Scholar] [CrossRef]
  79. Siegel, D.; Martin, T.; Nooka, A.; Harvey, R.D.; Vij, R.; Niesvizky, R.; Badros, A.Z.; Jagannath, S.; McCulloch, L.; Rajangam, K.; et al. Integrated safety profile of single-agent carfilzomib: Experience from 526 patients enrolled in 4 phase II clinical studies. Haematologica 2013, 98, 1753–1761. [Google Scholar] [CrossRef]
  80. Bishnoi, R.; Xie, Z.; Shah, C.; Bian, J.; Murthy, H.S.; Wingard, J.R.; Farhadfar, N. Real-world experience of carfilzomib-associated cardiovascular adverse events: SEER-Medicare data set analysis. Cancer Med. 2021, 10, 70–78. [Google Scholar] [CrossRef]
  81. Waxman, A.J.; Clasen, S.; Hwang, W.T.; Garfall, A.; Vogl, D.T.; Carver, J.; O’Quinn, R.; Cohen, A.D.; Stadtmauer, E.A.; Ky, B.; et al. Carfilzomib-Associated Cardiovascular Adverse Events: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, e174519. [Google Scholar] [CrossRef]
  82. Burger, J.A.; Tedeschi, A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Bairey, O.; Hillmen, P.; Barlett, N.L.; Li, J.; et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2015, 373, 2425–2437. [Google Scholar] [CrossRef] [PubMed]
  83. Dreyling, M.; Jurczak, W.; Jerkeman, M.; Silva, R.S.; Rusconi, C.; Trneny, M.; Offner, F.; Caballero, D.; Joao, C.; Witzens-Harig, M.; et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: An international, randomised, open-label, phase 3 study. Lancet 2016, 387, 770–778. [Google Scholar] [CrossRef] [PubMed]
  84. Owen, R.G.; McCarthy, H.; Rule, S.; D’Sa, S.; Thomas, S.K.; Tournilhac, O.; Forconi, F.; Kersten, M.J.; Zinzani, P.L.; Iyengar, S.; et al. Acalabrutinib monotherapy in patients with Waldenström macroglobulinemia: A single-arm, multicentre, phase 2 study. Lancet Haematol. 2020, 7, e112–e121. [Google Scholar] [CrossRef]
  85. Chen, S.T.; Azali, L.; Rosen, L.; Zhao, Q.; Wiczer, T.; Palettas, M.; Gambril, J.; Kola-Kehinde, O.; Ruz, P.; Kalathoor, S.; et al. Hypertension and incident cardiovascular events after next-generation BTKi therapy initiation. J. Hematol. Oncol. 2022, 15, 92. [Google Scholar] [CrossRef]
  86. Tam, C.S.; Opat, S.; D’Sa, S.; Jurczak, W.; Lee, H.P.; Cull, G.; Owen, R.G.; Marlton, P.; Wahlin, B.E.; Garcia Sanz, R.; et al. A randomized phase 3 trial of zanubrutinib vs ibrutinib in symptomatic Waldenström macroglobulinemia: The ASPEN study. Blood 2020, 136, 2038–2050. [Google Scholar] [CrossRef] [PubMed]
  87. Caldeira, D.; Alves, D.; Costa, J.; Ferreira, J.J.; Pinto, F.J. Ibrutinib increases the risk of hypertension and atrial fibrillation: Systematic review and meta-analysis. PLoS ONE 2019, 14, e0211228. [Google Scholar] [CrossRef]
  88. Dickerson, T.; Wiczer, T.; Waller, A.; Philippon, J.; Porter, K.; Haddad, D.; Guha, A.; Rogers, K.A.; Bhat, S.; Byrd, J.C.; et al. Hypertension and incident cardiovascular events following ibrutinib initiation. Blood 2019, 134, 1919–1928. [Google Scholar] [CrossRef] [PubMed]
  89. Ping, L.; Ding, N.; Shi, Y.; Feng, L.; Li, J.; Liu, Y.; Lin, Y.; Shi, C.; Wang, X.; Pan, Z.; et al. The Bruton’s tyrosine kinase inhibitor ibrutinib exerts immunomodulatory effects through regulation of tumor-infiltrating macrophages. Oncotarget 2017, 8, 39218–39229. [Google Scholar] [CrossRef]
  90. Kappers, M.H.; van Esch, J.H.; Sluiter, W.; Sleijfer, S.; Danser, A.H.; van den Meiracker, A.H. Hypertension induced by the tyrosine kinase inhibitor sunitinib is associated with increased circulating endothelin-1 levels. Hypertension 2010, 56, 675–681. [Google Scholar] [CrossRef]
  91. Wu, S.; Chen, J.J.; Kudelka, A.; Lu, J.; Zhu, X. Incidence and risk of hypertension with sorafenib in patients with cancer: A systematic review and meta-analysis. Lancet Oncol. 2008, 9, 117–123. [Google Scholar] [CrossRef]
  92. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.J.; et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N. Engl. J. Med. 2014, 371, 1877–1888. [Google Scholar] [CrossRef]
  93. Bronte, E.; Bronte, G.; Novo, G.; Rinaldi, G.; Bronte, F.; Passiglia, F.; Russo, A. Cardiotoxicity mechanisms of the combination of BRAF-inhibitors and MEK-inhibitors. Pharmacol. Ther. 2018, 192, 65–73. [Google Scholar] [CrossRef]
  94. Glen, C.; Tan, Y.Y.; Waterston, A.; Evans, T.R.J.; Jones, R.J.; Petrie, M.C.; Lang, N.N. Mechanistic and Clinical Overview Cardiovascular Toxicity of BRAF and MEK Inhibitors: JACC: CardioOncology State-of-the-Art Review. JACC Cardio Oncol. 2022, 4, 1–18. [Google Scholar] [CrossRef] [PubMed]
  95. Pandey, S.; Kashfi, S.; Hong, S.; Kalaria, A.; Kim, A.S. Onco-Hypertension in Patients with Kidney Disease. Am. J. Nephrol. 2024, 1–14. [Google Scholar] [CrossRef] [PubMed]
  96. Mincu, R.I.; Mahabadi, A.A.; Michel, L.; Mrotzek, S.M.; Schadendorf, D.; Rassaf, T.; Totzeck, M. Cardiovascular Adverse Events Associated With BRAF and MEK Inhibitors: A Systematic Review and Meta-analysis. JAMA Netw. Open 2019, 2, e198890. [Google Scholar] [CrossRef] [PubMed]
  97. Guha, A.; Jain, P.; Fradley, M.G.; Lenihan, D.; Gutierrez, J.M.; Jain, C.; de Lima, M.; Barnholtz-Sloan, J.S.; Oliveira, G.H.; Dowlati, A.; et al. Cardiovascular adverse events associated with BRAF versus BRAF/MEK inhibitor: Cross-sectional and longitudinal analysis using two large national registries. Cancer Med. 2021, 10, 3862–3872. [Google Scholar] [CrossRef]
  98. Larkin, J.; Del Vecchio, M.; Ascierto, P.A.; Krajsova, I.; Schachter, J.; Neyns, B.; Espinosa, E.; Garbe, C.; Chiaron Sileni, V.; Gogas, H.; et al. Vemurafenib in patients with BRAF(V600) mutated metastatic melanoma: An open-label, multicentre, safety study. Lancet Oncol. 2014, 15, 436–444. [Google Scholar] [CrossRef]
  99. Pandey, S.; Kalaria, A.; Jhaveri, K.D.; Herrmann, S.M.; Kim, A.S. Management of hypertension in patients with cancer: Challenges and considerations. Clin. Kidney J. 2023, 16, 2336–2348. [Google Scholar] [CrossRef]
  100. Sharabi, Y.; Dendi, R.; Holmes, C.; Goldstein, D.S. Baroreflex failure as a late sequela of neck irradiation. Hypertension 2003, 42, 110–116. [Google Scholar] [CrossRef]
  101. Timmers, H.J.; Wieling, W.; Karemaker, J.M.; Lenders, J.W. Cardiovascular responses to stress after carotid baroreceptor denervation in humans. Ann. N. Y. Acad. Sci. 2004, 1018, 515–519. [Google Scholar] [CrossRef]
  102. Piani, F.; Landolfo, M.; Fiorini, G.; D’Addato, S.; Mancia, G.; Borghi, C. Severe impaired blood pressure control caused by baroreflex failure as a late sequela of neck irradiation. J. Hypertens. 2020, 38, 553–556. [Google Scholar] [CrossRef] [PubMed]
  103. Klaus, R.; Niyazi, M.; Lange-Sperandio, B. Radiation-induced kidney toxicity: Molecular and cellular pathogenesis. Radiat. Oncol. 2021, 16, 43. [Google Scholar] [CrossRef]
  104. Cohen, E.P.; Lawton, C.A.; Moulder, J.E.; Becker, C.G.; Ash, R.C. Clinical course of late-onset bone marrow transplant nephropathy. Nephron 1993, 64, 626–635. [Google Scholar] [CrossRef]
  105. Cohen, E.P. Radiation nephropathy after bone marrow transplantation. Kidney Int. 2000, 58, 903–918. [Google Scholar] [CrossRef] [PubMed]
  106. Parker, G.A.; Cohen, E.P.; Li, N.; Takayama, K.; Farese, A.M.; MacVittie, T.J. Radiation Nephropathy in a Nonhuman Primate Model of Partial-Body Irradiation With Minimal Bone Marrow Sparing-Part 2: Histopathology, Mediators, and Mechanisms. Health Phys. 2019, 116, 409–425. [Google Scholar] [CrossRef] [PubMed]
  107. Baradaran-Ghahfarokhi, M. Radiation-induced kidney injury. J. Ren. Inj. Prev. 2012, 1, 49–50. [Google Scholar]
  108. Saka, B.; Bilge, A.K.; Umman, B.; Yilmaz, E.; Nisanci, Y.; Erten, N.; Karan, M.A.; Tascioglu, C. Bilateral renal artery stenosis after abdominal radiotherapy for Hodgkin’s disease. Int. J. Clin. Pract. 2003, 57, 247–248. [Google Scholar] [CrossRef]
  109. Bali, L.; Silhol, F.; Kateb, A.; Vaisse, B. Renal artery stenosis after abdominal radiotherapy. Ann. Cardiol. Angeiol. 2009, 58, 183–186. [Google Scholar] [CrossRef]
  110. Karati, D.; Mahadik, K.R.; Trivedi, P.; Kumar, D. Alkylating Agents, the Road Less Traversed, Changing Anticancer Therapy. Anticancer. Agents Med. Chem. 2022, 22, 1478–1495. [Google Scholar]
  111. Lajous, H.; Lelièvre, B.; Vauléon, E.; Lecomte, P.; Garcion, E. Rethinking Alkylating(-Like) Agents for Solid Tumor Management. Trends Pharmacol. Sci. 2019, 40, 342–357. [Google Scholar] [CrossRef]
  112. Zhu, X.; Lei, J.; Chen, R.; Chen, Z.; Xiong, Z.; Yang, L.; Jiang, M.; Zhang, H. Cancer Recurrence Fear and Return to Work in Breast Cancer Survivors: The Mediating Effects of Health Literacy. J. Multidiscip. Healthc. 2025, 18, 1031–1041. [Google Scholar] [CrossRef] [PubMed]
  113. Kooijmans, E.C.; Bökenkamp, A.; Tjahjadi, N.S.; Tettero, J.M.; van Dulmen-den Broeder, E.; van der Pal, H.J.; Veening, M.A. Early and late adverse renal effects after potentially nephrotoxic treatment for childhood cancer. Cochrane Database Syst. Rev. 2019, 3, Cd008944. [Google Scholar] [CrossRef] [PubMed]
  114. Soultati, A.; Mountzios, G.; Avgerinou, C.; Papaxoinis, G.; Pectasides, D.; Dimopoulos, M.A.; Papadimitriou, C. Endothelial vascular toxicity from chemotherapeutic agents: Preclinical evidence and clinical implications. Cancer Treat. Rev. 2012, 38, 473–483. [Google Scholar] [CrossRef]
  115. Nuver, J.; Smit, A.J.; Sleijfer, D.T.; van Gessel, A.I.; van Roon, A.M.; van der Meer, J.; van den Berg, M.P.; Burgerhof, J.G.M.; Hoekstra, H.J.; Sluiter, W.J.; et al. Microalbuminuria, decreased fibrinolysis, and inflammation as early signs of atherosclerosis in long-term survivors of disseminated testicular cancer. Eur. J. Cancer 2004, 40, 701–706. [Google Scholar] [CrossRef] [PubMed]
  116. Valentová, M.; Mladosievicová, B. Coronary heart disease and hypertension as late effects of testicular cancer treatment—A minireview. Klin. Onkol. 2011, 24, 18–22. [Google Scholar]
  117. Knijnenburg, S.L.; Jaspers, M.W.; van der Pal, H.J.; Schouten-van Meeteren, A.Y.; Bouts, A.H.; Lieverst, J.A.; Bökenkamp, A.; Koning, C.C.E.; Oldenburger, F.; Wilde, J.C.H.; et al. Renal dysfunction and elevated blood pressure in long-term childhood cancer survivors. Clin. J. Am. Soc. Nephrol. 2012, 7, 1416–1427. [Google Scholar] [CrossRef]
  118. Cohen, J.B.; Geara, A.S.; Hogan, J.J.; Townsend, R.R. Hypertension in Cancer Patients and Survivors: Epidemiology, Diagnosis, and Management. JACC Cardio Oncol. 2019, 1, 238–251. [Google Scholar] [CrossRef]
  119. Katsi, V.; Magkas, N.; Georgiopoulos, G.; Athanasiadi, E.; Virdis, A.; Masi, S.; Kliridis, P.; Hatziyanni, A.; Tsioufis, C.; Tousoulis, D. Arterial hypertension in patients under antineoplastic therapy: A systematic review. J. Hypertens. 2019, 37, 884–901. [Google Scholar] [CrossRef]
  120. Herradón, E.; González, C.; Uranga, J.A.; Abalo, R.; Martín, M.I.; López-Miranda, V. Characterization of Cardiovascular Alterations Induced by Different Chronic Cisplatin Treatments. Front. Pharmacol. 2017, 8, 196. [Google Scholar] [CrossRef]
  121. Rahman, A.A.; Stojanovska, V.; Pilowsky, P.; Nurgali, K. Platinum accumulation in the brain and alteration in the central regulation of cardiovascular and respiratory functions in oxaliplatin-treated rats. Pflug. Arch. 2021, 473, 107–120. [Google Scholar] [CrossRef]
  122. Sagstuen, H.; Aass, N.; Fosså, S.D.; Dahl, O.; Klepp, O.; Wist, E.A.; Wilsgaard, T.; Bremnes, R.M. Blood pressure and body mass index in long-term survivors of testicular cancer. J. Clin. Oncol. 2005, 23, 4980–4990. [Google Scholar] [CrossRef] [PubMed]
  123. Rix, U.; Hantschel, O.; Dürnberger, G.; Remsing Rix, L.L.; Planyavsky, M.; Fernbach, N.V.; Kaupe, I.; Bennett, K.L.; Valent, P.; Colinge, J.; et al. Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 2007, 110, 4055–4063. [Google Scholar] [CrossRef] [PubMed]
  124. Mulas, O.; Caocci, G.; Mola, B.; La Nasa, G. Arterial Hypertension and Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2021, 12, 674748. [Google Scholar] [CrossRef]
  125. Ai, N.; Chong, C.M.; Chen, W.; Hu, Z.; Su, H.; Chen, G.; Lei Wong, Q.W.; Ge, W. Ponatinib exerts anti-angiogenic effects in the zebrafish and human umbilical vein endothelial cells via blocking VEGFR signaling pathway. Oncotarget 2018, 9, 31958–31970. [Google Scholar] [CrossRef]
  126. Hadzijusufovic, E.; Albrecht-Schgoer, K.; Huber, K.; Hoermann, G.; Grebien, F.; Eisenwort, G.; Schgoer, W.; Herndlhofer, S.; Kaun, C.; Theurl, M.; et al. Nilotinib-induced vasculopathy: Identification of vascular endothelial cells as a primary target site. Leukemia 2017, 31, 2388–2397. [Google Scholar] [CrossRef]
  127. Chiarini, F.; Evangelisti, C.; McCubrey, J.A.; Martelli, A.M. Current treatment strategies for inhibiting mTOR in cancer. Trends Pharmacol. Sci. 2015, 36, 124–135. [Google Scholar] [CrossRef] [PubMed]
  128. Gasparri, M.L.; Bardhi, E.; Ruscito, I.; Papadia, A.; Farooqi, A.A.; Marchetti, C.; Bogani, G.; Ceccacci, I.; Mueller, M.D.; Benedetti Panici, P. PI3K/AKT/mTOR Pathway in Ovarian Cancer Treatment: Are We on the Right Track? Geburtshilfe Frauenheilkd 2017, 77, 1095–1103. [Google Scholar] [CrossRef]
  129. Karar, J.; Maity, A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 2011, 4, 51. [Google Scholar] [CrossRef]
  130. Motzer, R.J.; Hutson, T.E.; Glen, H.; Michaelson, M.D.; Molina, A.; Eisen, T.; Jassem, J.; Zolnierek, J.; Maroto, J.P.; Mellado, B.; et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: A randomised, phase 2, open-label, multicentre trial. Lancet Oncol. 2015, 16, 1473–1482. [Google Scholar] [CrossRef]
  131. Kaplan, B.; Qazi, Y.; Wellen, J.R. Strategies for the management of adverse events associated with mTOR inhibitors. Transpl. Rev. 2014, 28, 126–133. [Google Scholar] [CrossRef]
  132. Motzer, R.J.; Escudier, B.; Oudard, S.; Hutson, T.E.; Porta, C.; Bracarda, S.; Grünwald, V.; Thompson, J.A.; Figlin, R.A.; Hollaender, N.; et al. Efficacy of everolimus in advanced renal cell carcinoma: A double-blind, randomised, placebo-controlled phase III trial. Lancet 2008, 372, 449–456. [Google Scholar] [CrossRef] [PubMed]
  133. Deedwania, P. Hypertension, dyslipidemia, and insulin resistance in patients with diabetes mellitus or the cardiometabolic syndrome: Benefits of vasodilating β-blockers. J. Clin. Hypertens. 2011, 13, 52–59. [Google Scholar] [CrossRef]
  134. Attard, G.; Reid, A.H.; Auchus, R.J.; Hughes, B.A.; Cassidy, A.M.; Thompson, E.; Babu Oommen, N.; Folkerd, E.; Dowsett, M.; Arlt, W.; et al. Clinical and biochemical consequences of CYP17A1 inhibition with abiraterone given with and without exogenous glucocorticoids in castrate men with advanced prostate cancer. J. Clin. Endocrinol. Metab. 2012, 97, 507–516. [Google Scholar] [CrossRef]
  135. Hussain, M.; Fizazi, K.; Saad, F.; Rathenborg, P.; Shore, N.; Ferreira, U.; Ivaschenko, P.; Demirhan, E.; Modelska, K.; Phung, D.; et al. Enzalutamide in Men with Nonmetastatic, Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2018, 378, 2465–2474. [Google Scholar] [CrossRef]
  136. Martinez-Martin, F.J.; Kuzior, A.; Hernandez-Lazaro, A.; de Leon-Durango, R.J.; Rios-Gomez, C.; Santana-Ojeda, B.; Perez-Rivero, J.M.; Fernandez-Trujillo-Comenge, P.M.; Gonzalez-Diaz, P.; Arnas-Leon, C.; et al. Incidence of hypertension in young transgender people after a 5-year follow-up: Association with gender-affirming hormonal therapy. Hypertens. Res. 2023, 46, 219–225. [Google Scholar] [CrossRef]
  137. Tonia, T.; Mettler, A.; Robert, N.; Schwarzer, G.; Seidenfeld, J.; Weingart, O.; Hyde, C.; Engert, A.; Bohlius, J. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst. Rev. 2012, 12, Cd003407. [Google Scholar] [CrossRef]
  138. Pufall, M.A. Glucocorticoids and Cancer. Adv. Exp. Med. Biol. 2015, 872, 315–333. [Google Scholar] [PubMed]
  139. Vaziri, N.D. Mechanism of erythropoietin-induced hypertension. Am. J. Kidney Dis. 1999, 33, 821–828. [Google Scholar] [CrossRef] [PubMed]
  140. Vaziri, N.D.; Zhou, X.J.; Naqvi, F.; Smith, J.; Oveisi, F.; Wang, Z.Q.; Purdy, R.E. Role of nitric oxide resistance in erythropoietin-induced hypertension in rats with chronic renal failure. Am. J. Physiol. 1996, 271, E113–E122. [Google Scholar] [CrossRef]
  141. Goodwin, J.E.; Geller, D.S. Glucocorticoid-induced hypertension. Pediatr. Nephrol. 2012, 27, 1059–1066. [Google Scholar] [CrossRef]
  142. Baid, S.; Nieman, L.K. Glucocorticoid excess and hypertension. Curr. Hypertens. Rep. 2004, 6, 493–499. [Google Scholar] [CrossRef] [PubMed]
  143. Johnson, A.G.; Nguyen, T.V.; Day, R.O. Do nonsteroidal anti-inflammatory drugs affect blood pressure? A meta-analysis. Ann. Intern. Med. 1994, 121, 289–300. [Google Scholar] [CrossRef] [PubMed]
  144. Souza, V.B.; Silva, E.N.; Ribeiro, M.L.; Martins Wde, A. Hypertension in patients with cancer. Arq. Bras. Cardiol. 2015, 104, 246–252. [Google Scholar] [CrossRef]
  145. Hoorn, E.J.; Walsh, S.B.; McCormick, J.A.; Zietse, R.; Unwin, R.J.; Ellison, D.H. Pathogenesis of calcineurin inhibitor-induced hypertension. J. Nephrol. 2012, 25, 269–275. [Google Scholar] [CrossRef]
  146. Divac, N.; Naumović, R.; Stojanović, R.; Prostran, M. The Role of Immunosuppressive Medications in the Pathogenesis of Hypertension and Efficacy and Safety of Antihypertensive Agents in Kidney Transplant Recipients. Curr. Med. Chem. 2016, 23, 1941–1952. [Google Scholar] [CrossRef] [PubMed]
  147. Morales, J.M. Influence of the new immunosuppressive combinations on arterial hypertension after renal transplantation. Kidney Int. Suppl. 2002, 82, S81–S87. [Google Scholar] [CrossRef]
  148. Bursztyn, M.; Zelig, O.; Or, R.; Nagler, A. Isradipine for the prevention of cyclosporine-induced hypertension in allogeneic bone marrow transplant recipients: A randomized, double-blind study. Transplantation 1997, 63, 1034–1036. [Google Scholar] [CrossRef]
  149. Wang, J.; Wang, Z.; Zhao, Y. Incidence and risk of hypertension with ramucirumab in cancer patients: A meta-analysis of published studies. Clin. Drug Investig. 2015, 35, 221–228. [Google Scholar] [CrossRef]
  150. Zhao, T.; Wang, X.; Xu, T.; Xu, X.; Liu, Z. Bevacizumab significantly increases the risks of hypertension and proteinuria in cancer patients: A systematic review and comprehensive meta-analysis. Oncotarget 2017, 8, 51492–51506. [Google Scholar] [CrossRef]
  151. Ge, P.; Han, C.; Reyila, A.; Liu, D.; Hong, W.; Liu, J.; Zhang, J.; Han, X.; Li, X.; Huang, M.; et al. Risk of antiangiogenic adverse events in metastatic colorectal cancer patients receiving aflibercept in combination with chemotherapy: A meta-analysis. Medicine 2023, 102, e34793. [Google Scholar] [CrossRef]
  152. Qi, W.X.; Lin, F.; Sun, Y.J.; Tang, L.N.; He, A.N.; Yao, Y.; Shen, Z. Incidence and risk of hypertension with pazopanib in patients with cancer: A meta-analysis. Cancer Chemother. Pharmacol. 2013, 71, 431–439. [Google Scholar] [CrossRef] [PubMed]
  153. PARAPLATIN (Carboplatin) [Prescribing Information]: Food and Drug Administration. 2022. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=f88e0483-09be-4f60-be8f-964c02e3ece0 (accessed on 1 June 2025).
  154. TORISEL (Temsirolimus) [Prescribing Information]: Food and Drug Administration. 2025. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=95b7dc92-2180-42f1-8699-3c28f609e674 (accessed on 1 June 2025).
  155. Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615. [Google Scholar] [CrossRef]
  156. Furman, R.R.; Byrd, J.C.; Owen, R.G.; O’Brien, S.M.; Brown, J.R.; Hillmen, P.; Stephens, D.M.; Chernyukhin, N.; Lezhava, T.; Hamdy, A.M.; et al. Pooled analysis of safety data from clinical trials evaluating acalabrutinib monotherapy in mature B-cell malignancies. Leukemia 2021, 35, 3201–3211. [Google Scholar] [CrossRef] [PubMed]
  157. DANZITEN (Nilotinib) [Prescribing Information]: Food and Drug Administration. 2024. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=d288d165-3505-49bb-9b2e-124490d65f49 (accessed on 1 June 2025).
  158. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E., Jr.; Collins, K.J.; Dennison Himmelfarb, C.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 2018, 71, 1269–1324. [Google Scholar] [PubMed]
  159. Kallioinen, N.; Hill, A.; Horswill, M.S.; Ward, H.E.; Watson, M.O. Sources of inaccuracy in the measurement of adult patients’ resting blood pressure in clinical settings: A systematic review. J. Hypertens. 2017, 35, 421–441. [Google Scholar] [CrossRef]
  160. Johnson, K.C.; Whelton, P.K.; Cushman, W.C.; Cutler, J.A.; Evans, G.W.; Snyder, J.K.; Ambrosius, W.T.; Beddhu, S.; Cheung, A.K.; Fine, L.J.; et al. Blood Pressure Measurement in SPRINT (Systolic Blood Pressure Intervention Trial). Hypertension 2018, 71, 848–857. [Google Scholar] [CrossRef]
  161. Kronish, I.M.; Kent, S.; Moise, N.; Shimbo, D.; Safford, M.M.; Kynerd, R.E.; O’Beirne, R.; Sullivan, A.; Muntner, P. Barriers to conducting ambulatory and home blood pressure monitoring during hypertension screening in the United States. J. Am. Soc. Hypertens. 2017, 11, 573–580. [Google Scholar] [CrossRef] [PubMed]
  162. McManus, R.J.; Mant, J.; Haque, M.S.; Bray, E.P.; Bryan, S.; Greenfield, S.M.; Jones, M.I.; Jowett, S.; Little, P.; Penaloza, C.; et al. Effect of self-monitoring and medication self-titration on systolic blood pressure in hypertensive patients at high risk of cardiovascular disease: The TASMIN-SR randomized clinical trial. JAMA 2014, 312, 799–808. [Google Scholar] [CrossRef]
  163. Williams, B.; Mancia, G.; Spiering, W.; Rosei, E.A.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. The Task Force for the management of arterial hypertension of the European Society of Cardiology (ESC) and the European Society of Hypertension (ESH). G. Ital. Cardiol. 2018, 19, 3s–73s. [Google Scholar]
  164. Rao, V.U.; Reeves, D.J.; Chugh, A.R.; O’Quinn, R.; Fradley, M.G.; Raghavendra, M.; Dent, S.; Barac, A.; Lenihan, D. Clinical Approach to Cardiovascular Toxicity of Oral Antineoplastic Agents: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 77, 2693–2716. [Google Scholar] [CrossRef]
  165. Maitland, M.L.; Bakris, G.L.; Black, H.R.; Chen, H.X.; Durand, J.B.; Elliott, W.J.; Ivy, S.P.; Leier, C.V.; Lindfield, J.; Liu, G.; et al. Initial assessment, surveillance, and management of blood pressure in patients receiving vascular endothelial growth factor signaling pathway inhibitors. J. Natl. Cancer Inst. 2010, 102, 596–604. [Google Scholar] [CrossRef] [PubMed]
  166. Sahni, G. Onco-Hypertension: Changing Paradigm of Treating Hypertension in Patients With Cancer. J. Clin. Oncol. 2023, 41, 958–963. [Google Scholar] [CrossRef]
  167. Zhu, X.; Wu, S. Risks and management of hypertension in cancer patients undergoing targeted therapy: A review. Clin. Hypertens. 2022, 28, 14. [Google Scholar] [CrossRef]
  168. van Dorst, D.C.H.; Dobbin, S.J.H.; Neves, K.B.; Herrmann, J.; Herrmann, S.M.; Versmissen, J.; Mathijssen, R.H.J.; Jan Danser, A.H.; Lang, N.N. Hypertension and Prohypertensive Antineoplastic Therapies in Cancer Patients. Circ. Res. 2021, 128, 1040–1061. [Google Scholar] [CrossRef] [PubMed]
  169. Ferrer, T.; Arín, R.M.; Casis, E.; Torres-Jacome, J.; Sanchez-Chapula, J.A.; Casis, O. Mechanisms responsible for the altered cardiac repolarization dispersion in experimental hypothyroidism. Acta Physiol. 2012, 204, 502–512. [Google Scholar] [CrossRef]
  170. Beavers, C.J.; Rodgers, J.E.; Bagnola, A.J.; Beckie, T.M.; Campia, U.; Di Palo, K.E.; Okwuosa, T.M.; Przespolewski, E.R.; Dent, S.; American Heart Association Clinical Pharmacology Committee; et al. Cardio-Oncology Drug Interactions: A Scientific Statement From the American Heart Association. Circulation 2022, 145, e811–e838. [Google Scholar] [CrossRef] [PubMed]
  171. Nazer, B.; Humphreys, B.D.; Moslehi, J. Effects of novel angiogenesis inhibitors for the treatment of cancer on the cardiovascular system: Focus on hypertension. Circulation 2011, 124, 1687–1691. [Google Scholar] [CrossRef]
  172. Asgharzadeh, F.; Geraylow, K.R.; Khazaei, M.; Nassiri, M.; Hassanian, S.M.; Ferns, G.A.; Avan, A. Angiotensin-converting Enzyme Inhibitors and Angiotensin Receptor Blockers as Potential Therapeutic Options for Pancreatic Cancer. Curr. Cancer Drug Targets 2022, 22, 785–795. [Google Scholar] [CrossRef] [PubMed]
  173. Rosenthal, T.; Gavras, I. Renin-Angiotensin Inhibition in Combating Malignancy: A Review. Anticancer. Res. 2019, 39, 4597–4602. [Google Scholar] [CrossRef]
  174. Waliany, S.; Sainani, K.L.; Park, L.S.; Zhang, C.A.; Srinivas, S.; Witteles, R.M. Increase in Blood Pressure Associated With Tyrosine Kinase Inhibitors Targeting Vascular Endothelial Growth Factor. JACC Cardio Oncol. 2019, 1, 24–36. [Google Scholar] [CrossRef]
  175. Philip, L.J.; Findlay, S.G.; Gill, J.H. Baseline blood pressure and development of cardiotoxicity in patients treated with anthracyclines: A systematic review. Int. J. Cardiol. Cardiovasc. Risk Prev. 2022, 15, 200153. [Google Scholar] [CrossRef] [PubMed]
  176. Perini, M.V.; Dmello, R.S.; Nero, T.L.; Chand, A.L. Evaluating the benefits of renin-angiotensin system inhibitors as cancer treatments. Pharmacol. Ther. 2020, 211, 107527. [Google Scholar] [CrossRef] [PubMed]
  177. Derosa, L.; Izzedine, H.; Albiges, L.; Escudier, B. Hypertension and Angiotensin System Inhibitors in Patients with Metastatic Renal Cell Carcinoma. Oncol. Rev. 2016, 10, 298. [Google Scholar] [CrossRef] [PubMed]
  178. Curwen, J.O.; Musgrove, H.L.; Kendrew, J.; Richmond, G.H.; Ogilvie, D.J.; Wedge, S.R. Inhibition of vascular endothelial growth factor-a signaling induces hypertension: Examining the effect of cediranib (recentin; AZD2171) treatment on blood pressure in rat and the use of concomitant antihypertensive therapy. Clin. Cancer Res. 2008, 14, 3124–3131. [Google Scholar] [CrossRef]
  179. Araújo, W.F.; Naves, M.A.; Ravanini, J.N.; Schor, N.; Teixeira, V.P. Renin-angiotensin system (RAS) blockade attenuates growth and metastatic potential of renal cell carcinoma in mice. Urol. Oncol. 2015, 33, 389.e1–389.e7. [Google Scholar] [CrossRef]
  180. McKay, R.R.; Rodriguez, G.E.; Lin, X.; Kaymakcalan, M.D.; Hamnvik, O.P.R.; Sabbisetti, V.S.; Bhatt, R.S.; Simantov, R.; Choueiri, T.K. Angiotensin system inhibitors and survival outcomes in patients with metastatic renal cell carcinoma. Clin. Cancer Res. 2015, 21, 2471–2479. [Google Scholar] [CrossRef]
  181. Cheung, K.S.; Chan, E.W.; Seto, W.K.; Wong, I.C.K.; Leung, W.K. ACE (Angiotensin-Converting Enzyme) Inhibitors/Angiotensin Receptor Blockers Are Associated With Lower Colorectal Cancer Risk: A Territory-Wide Study with Propensity Score Analysis. Hypertension 2020, 76, 968–975. [Google Scholar] [CrossRef]
  182. Mir, O.; Coriat, R.; Ropert, S.; Cabanes, L.; Blanchet, B.; Camps, S.; Billemont, B.; Knebelmann, B.; Goldwasser, F. Treatment of bevacizumab-induced hypertension by amlodipine. Investig. New Drugs 2012, 30, 702–707. [Google Scholar] [CrossRef]
  183. Armillotta, M.; Angeli, F.; Paolisso, P.; Belmonte, M.; Raschi, E.; Di Dalmazi, G.; Amicone, S.; Canton, L.; Fedele, D.; Suma, N.; et al. Cardiovascular therapeutic targets of sodium-glucose co-transporter 2 (SGLT2) inhibitors beyond heart failure. Pharmacol. Ther. 2025, 270, 108861. [Google Scholar] [CrossRef]
  184. Alshnbari, A.S.; Millar, S.A.; O’Sullivan, S.E.; Idris, I. Effect of sodium-glucose cotransporter-2 inhibitors on endothelial function: A systematic review of preclinical studies. Diabetes Ther. 2020, 11, 1947–1963. [Google Scholar] [CrossRef]
  185. Ren, C.; Sun, K.; Zhang, Y.; Hu, Y.; Hu, B.; Zhao, J.; He, Z.; Ding, R.; Wang, W.; Liang, C. Sodium-Glucose CoTransporter-2 Inhibitor Empagliflozin Ameliorates Sunitinib-Induced Cardiac Dysfunction via Regulation of AMPK-mTOR Signaling Pathway-Mediated Autophagy. Front. Pharmacol. 2021, 12, 664181. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of hypertension.
Figure 1. Mechanisms of hypertension.
Jcdd 12 00235 g001
Figure 2. Friendly Guide: Managing Hypertension with VSPIs.
Figure 2. Friendly Guide: Managing Hypertension with VSPIs.
Jcdd 12 00235 g002
Table 1. Classes of Antineoplastic Agents Associated with Hypertension: Mechanisms of Action, Representative Medications, Relative Incidence of All-Grade Hypertension, and Proposed Pathophysiological Mechanisms.
Table 1. Classes of Antineoplastic Agents Associated with Hypertension: Mechanisms of Action, Representative Medications, Relative Incidence of All-Grade Hypertension, and Proposed Pathophysiological Mechanisms.
Medication ClassMechanism of ActionExample MedicationsRelative Incidence of All-Grade HTN †Impaired NO HomeostasisVascular Endothelial DysfunctionCapillary RarefactionImpaired NatriuresisAngiotensin-Mediated VasoconstrictionDirect Vascular ToxicityAbnormal Vascular RemodelingOxidative StressImpaired VEGF SignalingRenal ToxicitySympathetic Dysfunction
VSPIDisruption of VEGF-mediated angiogenesisRamucirumab [149]
(anti-VEGF-R2 Ab )
++++ +
Bevacizumab [150]
(anti-VEGF-A Ab)
Aflibercept
[151]
(anti-VEGF-Trap ligand Ab)
↑↑
Pazopanib [152]
(tyrosine kinase inhibitor)
↑↑
Proteosome inhibitors [6,73,75,76,77]Inhibition of ubiquitin-proteosome cascadeCarfilzomib↑↑++ + +
Bortezomib
Alkylating agents [6,75,112,113,114,115]Impaired genome replication and transcription via DNA cross-linkingBusulfan↑↑++ +++++
Ifosfamide
Platinum-containing compounds [6,110,114,119,120,121,122,153]Impaired genome replication and transcription via DNA cross-linkingCisplatin↑↑↑++ + ++
Carboplatin
mTOR inhibitors [6,129,130,131,154]Disruption of cellular metabolism, growth, and proliferation via serine-threonine kinase inhibitionEverolimus ++ +
Temsirolimus
BRAF/MEK [92,93,94,95,96,155]MAPK pathway inhibitionEncorafenib/binimetinib+
Vemurafenib/cobimetinib
BTK inhibitors [88,89,90,156]Impeded B cell receptor signalingAcalabrutinib++ + +
Ibrutinib↑↑↑
BCR-ABL TK inhibitors ‡ [123,124,125,126,157]Inhibition of BCR-ABL TK-mediated effects upon cellular division and apoptosisPonatinib↑↑ + +
Nilotinib
VSPI = VEGF signaling pathway inhibitor; VEGF = vascular endothelial growth factor; Ab = antibody; HTN = hypertension; NO = nitric oxide; DNA = deoxyribonucleic acid; mTOR = mammalian target of rapamycin; BRAF = rapidly accelerated fibrosarcoma B-type; MEK = mitogen-activated extracellular signal-regulated kinase; MAPK = mitogen-activated protein kinase; BTK = Bruton’s tyrosine kinase; BCR-ALB = breakpoint cluster region–Abelson leukemia virus 1; TK = tyrosine kinase. † Relative incidence: ↑: <25%; ↑↑: 25–50%, ↑↑↑: 50–75%. + symbol indicates the involvement or contribution of a particular mechanism in the development of hypertension. ‡ mechanisms of BCR-ABL TK inhibitor-induced HTN is not fully characterized. + symbol determines possible mechanism of hypertension.
Table 2. Adjuvant and radiation therapies.
Table 2. Adjuvant and radiation therapies.
Medication ClassMechanism of ActionExample MedicationsMechanism of HTN
Adjuvant TherapiesCorticosteroidsPredominantly glucocorticoid activity Prednisone, dexamethasonePredominantly mineralocorticoid-induced water and Na retention
Calcineurin inhibitorsImpaired T cell activation via inhibition of transcription factorsCyclosporine, tacrolimusRAAS activation, sympathetic dysfunction, increased proximal tubule Na resorption (NCC-mediated), endothelial dysfunction (ET1 mediated), oxidative stress, impaired NO homeostasis
Erythropoiesis stimulating agentsIncreased erythrocyte productionEpoetin alfa, epoetin betaIncreased blood viscosity, arterial remodeling, impaired NO homeostasis
NSAIDsDecreased prostaglandin synthesis via inhibition of COX enzymesKetorolac, ibuprofenImpaired natriuresis
Radiation TherapyCellular apoptosisN/ACarotid baroreceptor dysfunction, radiation nephropathy, radiation-induced renal artery stenosis
HTN = hypertension; RAAS = renin–angiotensin–aldosterone system; NCC = sodium chloride cotransporter; ET1 = endothelin-1; NO = nitric oxide; NSAIDs = non-steroid anti-inflammatory drugs; COX = cyclooxygenase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arriola-Montenegro, J.; Roth, J.; Gonzalez Suarez, M.L. Navigating the Complexities of Cancer Treatment-Induced Hypertension. J. Cardiovasc. Dev. Dis. 2025, 12, 235. https://doi.org/10.3390/jcdd12060235

AMA Style

Arriola-Montenegro J, Roth J, Gonzalez Suarez ML. Navigating the Complexities of Cancer Treatment-Induced Hypertension. Journal of Cardiovascular Development and Disease. 2025; 12(6):235. https://doi.org/10.3390/jcdd12060235

Chicago/Turabian Style

Arriola-Montenegro, Jose, John Roth, and Maria L. Gonzalez Suarez. 2025. "Navigating the Complexities of Cancer Treatment-Induced Hypertension" Journal of Cardiovascular Development and Disease 12, no. 6: 235. https://doi.org/10.3390/jcdd12060235

APA Style

Arriola-Montenegro, J., Roth, J., & Gonzalez Suarez, M. L. (2025). Navigating the Complexities of Cancer Treatment-Induced Hypertension. Journal of Cardiovascular Development and Disease, 12(6), 235. https://doi.org/10.3390/jcdd12060235

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop